201
|
Diaz-Padilla I, Wilson MK, Clarke BA, Hirte HW, Welch SA, Mackay HJ, Biagi JJ, Reedijk M, Weberpals JI, Fleming GF, Wang L, Liu G, Zhou C, Blattler C, Ivy SP, Oza AM. A phase II study of single-agent RO4929097, a gamma-secretase inhibitor of Notch signaling, in patients with recurrent platinum-resistant epithelial ovarian cancer: A study of the Princess Margaret, Chicago and California phase II consortia. Gynecol Oncol 2015; 137:216-22. [PMID: 25769658 DOI: 10.1016/j.ygyno.2015.03.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/04/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE A phase II study was performed to evaluate the efficacy and safety of single-agent RO4929097 (a gamma-secretase inhibitor) in patients with recurrent platinum-resistant ovarian cancer. EXPERIMENTAL DESIGN Women with progressive platinum-resistant ovarian cancer treated with ≤2 chemotherapy regimens for recurrent disease were enrolled in this trial. Patients received oral RO4929097 at 20 mg once daily, 3 days on/4 days off each week in a three week cycle. The primary endpoint was progression-free survival (PFS) rate at the end of 4 cycles. Secondary objectives included assessment of the safety of RO4929097 and exploration of molecular correlates of outcome in archival tumor tissue and serum. RESULTS Of 45 patients enrolled, 40 were evaluable for response. Thirty-seven (82%) patients had high-grade ovarian cancer. No objective responses were observed. Fifteen patients (33%) had stable disease as their best response, with a median duration of 3.1 months. The median PFS for the whole group was 1.3 months (1.2-2.5). Treatment was generally well tolerated with 10% of patients discontinuing treatment due to an adverse event. In high grade serous ovarian cancer patients, the median PFS trended higher when the expression of intracellular Notch (NICD) protein by immunohistochemistry was high versus low (3.3 versus 1.3 months, p=0.09). No clear relationship between circulating angiogenic factors and PFS was found despite a suggestion of an improved outcome with higher baseline VEGFA levels. CONCLUSIONS RO4929097 has insufficient activity as a single-agent in platinum-resistant ovarian cancer to warrant further study as monotherapy. Future studies are needed to explore the potential for cohort enrichment using NICD expression.
Collapse
Affiliation(s)
- Ivan Diaz-Padilla
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Michelle K Wilson
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Blaise A Clarke
- Department of Laboratory Medicine, University of Toronto, Toronto, Canada
| | - Hal W Hirte
- Division of Medical Oncology, Juravinski Cancer Centre, Hamilton, Ontario, Canada
| | - Stephen A Welch
- Division of Medical Oncology, London Regional Cancer Program, London, Ontario, Canada
| | - Helen J Mackay
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Jim J Biagi
- Department of Oncology, Cancer Centre of Southeastern Ontario, Kingston, Ontario, Canada
| | - Michael Reedijk
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Johanne I Weberpals
- Division of Gynecologic Oncology, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Gini F Fleming
- The University of Chicago Medical Center, Chicago, IL, USA
| | - Lisa Wang
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Chen Zhou
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Chantale Blattler
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - S Percy Ivy
- National Cancer Institute, Division of Cancer Treatment and Diagnosis, Cancer Therapy Evaluation Program, Investigational Drug Branch, Rockville, MD, USA
| | - Amit M Oza
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
| |
Collapse
|
202
|
House CD, Hernandez L, Annunziata CM. In vitro enrichment of ovarian cancer tumor-initiating cells. J Vis Exp 2015. [PMID: 25742116 PMCID: PMC4354662 DOI: 10.3791/52446] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Evidence suggests that small subpopulations of tumor cells maintain a unique self-renewing and differentiation capacity and may be responsible for tumor initiation and/or relapse. Clarifying the mechanisms by which these tumor-initiating cells (TICs) support tumor formation and progression could lead to the development of clinically favorable therapies. Ovarian cancer is a heterogeneous and highly recurrent disease. Recent studies suggest TICs may play an important role in disease biology. We have identified culture conditions that enrich for TICs from ovarian cancer cell lines. Growing either adherent cells or non-adherent ‘floater’ cells in a low attachment plate with serum free media in the presence of growth factors supports the propagation of ovarian cancer TICs with stem cell markers (CD133 and ALDH activity) and increased tumorigenicity without the need to physically separate the TICs from other cell types within the culture. Although the presence of floater cells is not common for all cell lines, this population of cells with innate low adherence may have high tumorigenic potential.Compared to adherent cells grown in the presence of serum, TICs readily form spheres, are significantly more tumorigenic in mice, and express putative stem cell markers. The conditions are easy to establish in a timely manner and can be used to study signaling pathways important for maintaining stem characteristics, and to identify drugs or combinations of drugs targeting TICs. The culture conditions described herein are applicable for a variety of ovarian cancer cells of epithelial origin and will be critical in providing new information about the role of TICs in tumor initiation, progression, and relapse.
Collapse
|
203
|
OVCAR-3 spheroid-derived cells display distinct metabolic profiles. PLoS One 2015; 10:e0118262. [PMID: 25688563 PMCID: PMC4331360 DOI: 10.1371/journal.pone.0118262] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 01/07/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction Recently, multicellular spheroids were isolated from a well-established epithelial ovarian cancer cell line, OVCAR-3, and were propagated in vitro. These spheroid-derived cells displayed numerous hallmarks of cancer stem cells, which are chemo- and radioresistant cells thought to be a significant cause of cancer recurrence and resultant mortality. Gene set enrichment analysis of expression data from the OVCAR-3 cells and the spheroid-derived putative cancer stem cells identified several metabolic pathways enriched in differentially expressed genes. Before this, there had been little previous knowledge or investigation of systems-scale metabolic differences between cancer cells and cancer stem cells, and no knowledge of such differences in ovarian cancer stem cells. Methods To determine if there were substantial metabolic changes corresponding with these transcriptional differences, we used two-dimensional gas chromatography coupled to mass spectrometry to measure the metabolite profiles of the two cell lines. Results These two cell lines exhibited significant metabolic differences in both intracellular and extracellular metabolite measurements. Principal components analysis, an unsupervised dimensional reduction technique, showed complete separation between the two cell types based on their metabolite profiles. Pathway analysis of intracellular metabolomics data revealed close overlap with metabolic pathways identified from gene expression data, with four out of six pathways found enriched in gene-level analysis also enriched in metabolite-level analysis. Some of those pathways contained multiple metabolites that were individually statistically significantly different between the two cell lines, with one of the most broadly and consistently different pathways, arginine and proline metabolism, suggesting an interesting hypothesis about cancerous and stem-like metabolic phenotypes in this pair of cell lines. Conclusions Overall, we demonstrate for the first time that metabolism in an ovarian cancer stem cell line is distinct from that of more differentiated isogenic cancer cells, supporting the potential importance of metabolism in the differences between cancer cells and cancer stem cells.
Collapse
|
204
|
Zhang Q, Cai DJ, Li B. Ovarian cancer stem-like cells elicit the polarization of M2 macrophages. Mol Med Rep 2015; 11:4685-93. [PMID: 25672286 DOI: 10.3892/mmr.2015.3323] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 12/19/2014] [Indexed: 11/05/2022] Open
Abstract
Ovarian cancer is a life‑threatening disease in females worldwide. The polarization of macrophages is crucial in oncogenesis and the development of ovarian cancer. Increasing evidence has supported the correlation between ovarian cancer stem‑like cells (OCSCs) and macrophages, however, whether OCSCs can affect the polarization of macrophages and the underlying mechanisms involved remain to be elucidated. To examine the interplay between OCSCs and macrophages, a co‑culture system was used to detect the effect of OCSCs on macrophage polarization. The expression of cluster of differentiation 206+ and the secretion of interleukin‑10 were significantly increased and the production of tumor necrosis factor‑α was suppressed, confirming macrophage polarization to M2 macrophages. Further investigation of the macrophages in a Transwell culture system with OCSCs revealed polarization to the M2 macrophages to a similar extent, indicating that the cytokines of the OCSCs, rather than direct cell‑cell contact, are important for the polarization of M2 macrophages. Furthermore, the expression levels of chemokine (C‑C motif) ligand (CCL)2, cyclooxygenase (COX)‑2 and prostaglandin E2 (PGE2) were increased in the Transwell system and the inhibition of COX‑2, but not CCL2, significantly decreased the polarization of the M2 macrophages. In addition, mechanistic analysis revealed the importance of the COX‑2/PGE2 pathway in OCSCs to activate Janus kinase (JAK) signaling in macrophages to elicit M2 polarization. These findings provided the first evidence, to the best of our knowledge, that OCSCs are capable of altering macrophages into the M2 phenotype via the overexpression of COX‑2 and the increased production of PGE2 cytokines and that the JAK signaling pathway in macrophages is important for this alteration. The present study provided evidence supporting possible molecular targets for cancer treatment.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Da-Jun Cai
- Department of Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Bin Li
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
205
|
|
206
|
Qiao L, Liang N, Zhang J, Xie J, Liu F, Xu D, Yu X, Tian Y. Advanced research on vasculogenic mimicry in cancer. J Cell Mol Med 2015; 19:315-26. [PMID: 25598425 PMCID: PMC4407602 DOI: 10.1111/jcmm.12496] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/20/2014] [Indexed: 12/20/2022] Open
Abstract
Vasculogenic mimicry (VM) is a brand-new tumour vascular paradigm independent of angiogenesis that describes the specific capacity of aggressive cancer cells to form vessel-like networks that provide adequate blood supply for tumour growth. A variety of molecule mechanisms and signal pathways participate in VM induction. Additionally, cancer stem cell and epithelial-mesenchymal transitions are also shown to be implicated in VM formation. As a unique perfusion way, VM is associated with tumour invasion, metastasis and poor cancer patient prognosis. Due to VM's important effects on tumour progression, more VM-related strategies are being utilized for anticancer treatment. Here, with regard to the above aspects, we make a review of advanced research on VM in cancer.
Collapse
Affiliation(s)
- Lili Qiao
- Department of Oncology, Shandong University School of Medicine, Jinan, Shandong Pro, China
| | | | | | | | | | | | | | | |
Collapse
|
207
|
Martínez-Serrano MJ, Caballero-Baños M, Vilella R, Vidal L, Pahisa J, Martínez-Roman S. Is sphere assay useful for the identification of cancer initiating cells of the ovary? Int J Gynecol Cancer 2015; 25:12-7. [PMID: 25365589 DOI: 10.1097/igc.0000000000000320] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Current evidence suggests that the presence of tumor-initiating cells (TICs) in epithelial ovarian cancer (EOC) has a role in chemoresistance and relapse. Surface markers such as CD44(+)/CD24(-), CD117(+), and CD133(+) expression have been reported as potential markers for TICs related to ovarian cancer and tumorigenic cell lines. In this study, we have investigated if spheroid forms are TIC specific or whether they can also be produced by somatic stem cells from healthy tissue in vitro. In addition, we also investigated the specificity of surface markers to identify TICs from papillary serous EOC patients. METHODS Cells were obtained from fresh tumors from 10 chemotherapy-naive patients with EOC, and cells from ovarian and tubal epithelium were obtained from 5 healthy menopausal women undergoing surgery for benign pathology and cultured in standard and in selective medium. Cells forming nonadherent spheroids were considered TICs, and the adherent cells were considered as non-TIC-like. Percentages of CD24(+), CD44(+), CD117(+), CD133(+), and vascular endothelial growth factor receptor (VEGF-R)(+) cell surface markers were analyzed by flow cytometry. RESULTS Four of 10 EOC cell tissues were excluded from the study. Tumor cells cultured in selective medium developed spheroid forms after 1 to 7 weeks in 5 of 6 EOC patients. No spheroid forms were observed in cultures of cells from healthy women. Unlike previously published data, low levels of CD24(+), CD44(+), CD117(+), and VEGF-R(+) expression were observed in spheroid cells, whereas expression of CD133(+) was moderate but higher in adherent cells from papillary serous EOC cells in comparison with adherent cells from controls. CONCLUSIONS Papillary serous EOC contains TICs that form spheroids with low expression of CD44(+), CD24(+), CD117(+) and VEGF-R(+). Further research is required to find specific surface markers to identify papillary serous TICs.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/metabolism
- Biomarkers, Tumor/metabolism
- Case-Control Studies
- Cells, Cultured
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- Female
- Flow Cytometry
- Follow-Up Studies
- Humans
- Middle Aged
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Neoplasm Staging
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Ovary/metabolism
- Ovary/pathology
- Pilot Projects
- Prognosis
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- María José Martínez-Serrano
- *Institut Clinic of Gynecology, Obstetrics and Neonatology, †Immunology Department, Centre de Diagnòstic Biomèdic, and ‡Institut Clinic of Hematologic Diseases and Clinical Oncology, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine-University of Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
208
|
Tong L, Yuan Y, Wu S. Therapeutic microRNAs targeting the NF-kappa B signaling circuits of cancers. Adv Drug Deliv Rev 2015; 81:1-15. [PMID: 25220353 DOI: 10.1016/j.addr.2014.09.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/27/2014] [Accepted: 09/03/2014] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) not only directly regulate NF-κB expression, but also up- or down-regulate NF-κB activity via upstream and downstream signaling pathways of NF-κB. In many cancer cells, miRNA expressions are altered accompanied with an elevation of NF-κB activity, which often plays a role in promoting cancer development and progression as well as hindering the effectiveness of chemo and radiation therapies. Thus NF-κB-targeting miRNAs have been identified and characterized as potential therapeutics for cancer treatment and sensitizers of chemo and radiotherapies. However, due to cross-targeting and instability of miRNAs, some limitations of using miRNA as cancer therapeutics still exist. In this review, the mechanisms for miRNA-mediated alteration of NF-κB expression and activation in different types of cancers will be discussed. The results of therapeutic use of NF-κB-targeting miRNA for cancer treatment will be examined. Some limitations, challenges and potential strategies in future development of miRNA as cancer therapeutics are also assessed.
Collapse
|
209
|
Smilax china L. rhizome extract inhibits nuclear factor-κB and induces apoptosis in ovarian cancer cells. Chin J Integr Med 2014; 21:907-15. [DOI: 10.1007/s11655-014-1788-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Indexed: 01/08/2023]
|
210
|
Identification of differently expressed genes with specific SNP Loci for breast cancer by the integration of SNP and gene expression profiling analyses. Pathol Oncol Res 2014; 21:469-75. [PMID: 25408372 DOI: 10.1007/s12253-014-9851-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 10/14/2014] [Indexed: 01/06/2023]
Abstract
This study aims to explore the relationship between gene polymorphism and breast cancer, and to screen DEGs (differentially expressed genes) with SNPs (single nucleotide polymorphisms) related to breast cancer. The SNPs of 17 patients and the preprocessed SNP profiling GSE 32258 (38 cases of normal breast cells) were combined to identify their correlation with breast cancer using chi-square test. The gene expression profiling batch8_9 (38 cases of patients and 8 cases of normal tissue) was preprocessed with limma package, and the DEGs were filtered out. Then fisher's method was applied to integrate DEGs and SNPs associated with breast cancer. With NetBox software, TRED (Transcriptional Regulatory Element Database) and UCSC (University of California Santa Cruz) database, genes-associated network and transcriptional regulatory network were constructed using cytoscape software. Further, GO (Gene Ontology) and KEGG analyses were performed for genes in the networks by using siggenes. In total, 332 DEGs were identified. There were 160 breast cancer-related SNPs related to 106 genes of gene expression profiling (19 were significant DEGs). Finally, 11co-correlated DEGs were selected. In genes-associated network, 9 significant DEGs were correlated to 23 LINKER genes while, in transcriptional regulatory network, E2F1 had regulatory relationships with 7 DEGs including MTUS1, CD44, CCNB1 and CCND2. KRAS with SNP locus of rs1137282 was involved in 35 KEGG pathways. The genes of MTUS1, CD44, CCNB1, CCND2 and KRAS with specific SNP loci may be used as biomarkers for diagnosis of breast cancer. Besides, E2F1 was recognized as the transcription factor of 7 DEGs including MTUS1, CD44, CCNB1 and CCND2.
Collapse
|
211
|
Ovarian cancer stem cells express ROR1, which can be targeted for anti-cancer-stem-cell therapy. Proc Natl Acad Sci U S A 2014; 111:17266-71. [PMID: 25411317 DOI: 10.1073/pnas.1419599111] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although initially responsive to chemotherapy, many patients with ovarian cancer subsequently develop relapsed and potentially fatal metastatic disease, which is thought to develop from cancer stem cells (CSCs) that are relatively resistant to conventional therapy. Here, we show that CSCs express a type I receptor tyrosine kinase-like orphan receptor (ROR1), which is expressed during embryogenesis and by many different cancers, but not normal postpartum tissues. Ovarian cancers with high levels of ROR1 had stem cell-like gene-expression signatures. Furthermore, patients with ovarian cancers with high levels of ROR1 had higher rates of relapse and a shorter median survival than patients with ovarian cancers that expressed low-to-negligible amounts of ROR1. We found that ROR1-positive (ROR1(+)) cells isolated from primary tumor-derived xenografts (PDXs) also expressed aldehyde dehydrogenase 1 (ALDH1) and had a greater capacity to form spheroids and to engraft immune-deficient mice than did ROR1-negative (ROR1(Neg)) ovarian cancer cells isolated from the same tumor population. Treatment with UC-961, an anti-ROR1 mAb, or shRNA silencing of ROR1 inhibited expression of the polycomb ring-finger oncogene, Bmi-1, and other genes associated with the epithelial-mesenchymal transition. Moreover, shRNA silencing of ROR1, depletion of ROR1(+) cells, or treatment with UC-961 impaired the capacity of ovarian cancer cells to form spheroids or tumor xenografts. More importantly, treatment with anti-ROR1 affected the capacity of the xenograft to reseed a virgin mouse, indicating that targeting ROR1 may affect CSC self-renewal. Collectively, these studies indicate that ovarian CSCs express ROR1, which contributes to their capacity to form tumors, making ROR1 a potential target for the therapy of patients with ovarian cancer.
Collapse
|
212
|
Liu KC, Yo YT, Huang RL, Wang YC, Liao YP, Huang TS, Chao TK, Lin CK, Weng SJ, Ma KH, Chang CC, Yu MH, Lai HC. Ovarian cancer stem-like cells show induced translineage-differentiation capacity and are suppressed by alkaline phosphatase inhibitor. Oncotarget 2014; 4:2366-82. [PMID: 24280306 PMCID: PMC3926833 DOI: 10.18632/oncotarget.1424] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Spheroid formation is one property of stem cells—such as embryo-derived or neural stem cells—that has been used for the enrichment of cancer stem-like cells (CSLCs). However, it is unclear whether CSLC-derived spheroids are heterogeneous or whether they share common embryonic stemness properties. Understanding these features might lead to novel therapeutic approaches. Ovarian carcinoma is a deadly disease of women. We identified two types of spheroids (SR1 and SR2) from ovarian cancer cell lines and patients' specimens according to their morphology. Both types expressed stemness markers and could self-renew and initiate tumors when a low number of cells were used. Only SR1 could differentiate into multiple-lineage cell types under specific induction conditions. SR1 spheroids could differentiate to SR2 spheroids through epithelial–mesenchymal transition. Alkaline phosphatase (ALP) was highly expressed in SR1 spheroids, decreased in SR2 spheroids, and was absent in differentiated progenies in accordance with the loss of stemness properties. We verified that ALP can be a marker for ovarian CSLCs, and patients with greater ALP expression is related to advanced clinical stages and have a higher risk of recurrence and lower survival rate. The ALP inhibitor, levamisole, disrupted the self-renewal of ovarian CSLCs in vitro and tumor growth in vivo. In summary, this research provides a plastic ovarian cancer stem cell model and a new understanding of the cross-link between stem cells and cancers. This results show that ovarian CSLCs can be suppressed by levamisole. Our findings demonstrated that some ovarian CSLCs may restore ALP activity, and this suggests that inhibition of ALP activity may present a new opportunity for treatment of ovarian cancer.
Collapse
Affiliation(s)
- Kuei-Chun Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Sumi NJ, Lima E, Pizzonia J, Orton SP, Craveiro V, Joo W, Holmberg JC, Gurrea M, Yang-Hartwich Y, Alvero A, Mor G. Murine model for non-invasive imaging to detect and monitor ovarian cancer recurrence. J Vis Exp 2014:e51815. [PMID: 25407815 PMCID: PMC4353409 DOI: 10.3791/51815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer is the most lethal gynecologic malignancy in the United States. Although patients initially respond to the current standard of care consisting of surgical debulking and combination chemotherapy consisting of platinum and taxane compounds, almost 90% of patients recur within a few years. In these patients the development of chemoresistant disease limits the efficacy of currently available chemotherapy agents and therefore contributes to the high mortality. To discover novel therapy options that can target recurrent disease, appropriate animal models that closely mimic the clinical profile of patients with recurrent ovarian cancer are required. The challenge in monitoring intra-peritoneal (i.p.) disease limits the use of i.p. models and thus most xenografts are established subcutaneously. We have developed a sensitive optical imaging platform that allows the detection and anatomical location of i.p. tumor mass. The platform includes the use of optical reporters that extend from the visible light range to near infrared, which in combination with 2-dimensional X-ray co-registration can provide anatomical location of molecular signals. Detection is significantly improved by the use of a rotation system that drives the animal to multiple angular positions for 360 degree imaging, allowing the identification of tumors that are not visible in single orientation. This platform provides a unique model to non-invasively monitor tumor growth and evaluate the efficacy of new therapies for the prevention or treatment of recurrent ovarian cancer.
Collapse
Affiliation(s)
- Natalia J Sumi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine
| | - Eydis Lima
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine
| | | | | | - Vinicius Craveiro
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine
| | - Wonduk Joo
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine
| | - Jennie C Holmberg
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine
| | - Marta Gurrea
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine
| | - Ayesha Alvero
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine;
| | - Gil Mor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine
| |
Collapse
|
214
|
Flesken-Nikitin A, Odai-Afotey AA, Nikitin AY. Role of the stem cell niche in the pathogenesis of epithelial ovarian cancers. Mol Cell Oncol 2014; 1:e963435. [PMID: 27308341 PMCID: PMC4905019 DOI: 10.4161/23723548.2014.963435] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/02/2014] [Accepted: 08/07/2014] [Indexed: 01/10/2023]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related deaths among women in the United States. Recent extensive genomic analyses of epithelial ovarian cancer (EOC), particularly the most common and deadly form of high-grade serous ovarian carcinoma, have provided important insights into the repertoire of molecular aberrations that are characteristic for this malignancy. However, interpretation of the discovered aberrations is complicated because the origin and mechanisms of progression of EOC remain uncertain. Here, we summarize current views on the cell of origin of EOC and discuss recent findings of a cancer-prone stem cell niche for ovarian surface epithelium, one of the major likely sources of EOC. We also outline future directions and challenges in studying the role of stem cell niches in EOC pathogenesis.
Collapse
Affiliation(s)
- Andrea Flesken-Nikitin
- Department of Biomedical Sciences and Stem Cell Program; Cornell University ; Ithaca, NY USA
| | - Ashley A Odai-Afotey
- Department of Biomedical Sciences and Stem Cell Program; Cornell University ; Ithaca, NY USA
| | - Alexander Yu Nikitin
- Department of Biomedical Sciences and Stem Cell Program; Cornell University ; Ithaca, NY USA
| |
Collapse
|
215
|
Garson K, Vanderhyden BC. Epithelial ovarian cancer stem cells: underlying complexity of a simple paradigm. Reproduction 2014; 149:R59-70. [PMID: 25301968 DOI: 10.1530/rep-14-0234] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The lack of significant progress in the treatment of epithelial ovarian cancer (EOC) underscores the need to gain a better understanding of the processes that lead to chemoresistance and recurrence. The cancer stem cell (CSC) hypothesis offers an attractive explanation of how a subpopulation of cells within a patient's tumour might remain refractory to treatment and subsequently form the basis of recurrent chemoresistant disease. This review examines the literature defining somatic stem cells of the ovary and fallopian tube, two tissues that give rise to EOC. In addition, considerable research has been reviewed, that has identified subpopulations of EOC cells, based on marker expression (CD133, CD44, CD117, CD24, epithelial cell adhesion molecule, LY6A, ALDH1 and side population (SP)), which are enriched for tumour initiating cells (TICs). While many studies identified either CD133 or CD44 as markers useful for enriching for TICs, there is little consensus. This suggests that EOC cells may have a phenotypic plasticity that may preclude the identification of universal markers defining a CSC. The assay that forms the basis of quantifying TICs is the xenograft assay. Considerable controversy surrounds the xenograft assay and it is essential that some of the potential limitations be examined in this review. Highlighting such limitations or weaknesses is required to properly evaluate data and broaden our interpretation of potential mechanisms that might be contributing to the pathogenesis of ovarian cancer.
Collapse
Affiliation(s)
- Kenneth Garson
- Ottawa Hospital Research InstituteCentre for Cancer Therapeutics, Ottawa, Ontario, Canada K1H 8L6Department of Cellular and Molecular MedicineFaculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Barbara C Vanderhyden
- Ottawa Hospital Research InstituteCentre for Cancer Therapeutics, Ottawa, Ontario, Canada K1H 8L6Department of Cellular and Molecular MedicineFaculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5 Ottawa Hospital Research InstituteCentre for Cancer Therapeutics, Ottawa, Ontario, Canada K1H 8L6Department of Cellular and Molecular MedicineFaculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| |
Collapse
|
216
|
Ahmed N, Abubaker K, Findlay JK. Ovarian cancer stem cells: Molecular concepts and relevance as therapeutic targets. Mol Aspects Med 2014; 39:110-25. [DOI: 10.1016/j.mam.2013.06.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/12/2013] [Accepted: 06/14/2013] [Indexed: 12/12/2022]
|
217
|
Mimeault M, Batra SK. Altered gene products involved in the malignant reprogramming of cancer stem/progenitor cells and multitargeted therapies. Mol Aspects Med 2014; 39:3-32. [PMID: 23994756 PMCID: PMC3938987 DOI: 10.1016/j.mam.2013.08.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 08/16/2013] [Accepted: 08/21/2013] [Indexed: 12/17/2022]
Abstract
Recent studies in the field of cancer stem cells have revealed that the alterations in key gene products involved in the epithelial-mesenchymal transition (EMT) program, altered metabolic pathways such as enhanced glycolysis, lipogenesis and/or autophagy and treatment resistance may occur in cancer stem/progenitor cells and their progenies during cancer progression. Particularly, the sustained activation of diverse developmental cascades such as hedgehog, epidermal growth factor receptor (EGFR), Wnt/β-catenin, Notch, transforming growth factor-β (TGF-β)/TGF-βR receptors and/or stromal cell-derived factor-1 (SDF-1)/CXC chemokine receptor 4 (CXCR4) can play critical functions for high self-renewal potential, survival, invasion and metastases of cancer stem/progenitor cells and their progenies. It has also been observed that cancer cells may be reprogrammed to re-express different pluripotency-associated stem cell-like markers such as Myc, Oct-3/4, Nanog and Sox-2 along the EMT process and under stressful and hypoxic conditions. Moreover, the enhanced expression and/or activities of some drug resistance-associated molecules such as Bcl-2, Akt/molecular target of rapamycin (mTOR), nuclear factor-kappaB (NF-κB), hypoxia-inducible factors (HIFs), macrophage inhibitory cytokine-1 (MIC-1) and ATP-binding cassette (ABC) multidrug transporters frequently occur in cancer cells during cancer progression and metastases. These molecular events may cooperate for the survival and acquisition of a more aggressive and migratory behavior by cancer stem/progenitor cells and their progenies during cancer transition to metastatic and recurrent disease states. Of therapeutic interest, these altered gene products may also be exploited as molecular biomarkers and therapeutic targets to develop novel multitargeted strategies for improving current cancer therapies and preventing disease relapse.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, College of Medicine, Fred & Pamela Buffett Cancer Center, Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, Fred & Pamela Buffett Cancer Center, Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| |
Collapse
|
218
|
p53 protein aggregation promotes platinum resistance in ovarian cancer. Oncogene 2014; 34:3605-16. [PMID: 25263447 DOI: 10.1038/onc.2014.296] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/25/2014] [Accepted: 07/31/2014] [Indexed: 12/24/2022]
Abstract
High-grade serous ovarian carcinoma (HGSOC), the most lethal gynecological cancer, often leads to chemoresistant diseases. The p53 protein is a key transcriptional factor regulating cellular homeostasis. A majority of HGSOCs have inactive p53 because of genetic mutations. However, genetic mutation is not the only cause of p53 inactivation. The aggregation of p53 protein has been discovered in different types of cancers and may be responsible for impairing the normal transcriptional activation and pro-apoptotic functions of p53. We demonstrated that in a unique population of HGSOC cancer cells with cancer stem cell properties, p53 protein aggregation is associated with p53 inactivation and platinum resistance. When these cancer stem cells differentiated into their chemosensitive progeny, they lost tumor-initiating capacity and p53 aggregates. In addition to the association of p53 aggregation and chemoresistance in HGSOC cells, we further demonstrated that the overexpression of a p53-positive regulator, p14ARF, inhibited MDM2-mediated p53 degradation and led to the imbalance of p53 turnover that promoted the formation of p53 aggregates. With in vitro and in vivo models, we demonstrated that the inhibition of p14ARF could suppress p53 aggregation and sensitize cancer cells to platinum treatment. Moreover, by two-dimensional gel electrophoresis and mass spectrometry we discovered that the aggregated p53 may function uniquely by interacting with proteins that are critical for cancer cell survival and tumor progression. Our findings help us understand the poor chemoresponse of a subset of HGSOC patients and suggest p53 aggregation as a new marker for chemoresistance. Our findings also suggest that inhibiting p53 aggregation can reactivate p53 pro-apoptotic function. Therefore, p53 aggregation is a potential therapeutic target for reversing chemoresistance. This is paramount for improving ovarian cancer patients' responses to chemotherapy, and thus increasing their survival rate.
Collapse
|
219
|
Kakar SS, Ratajczak MZ, Powell KS, Moghadamfalahi M, Miller DM, Batra SK, Singh SK. Withaferin a alone and in combination with cisplatin suppresses growth and metastasis of ovarian cancer by targeting putative cancer stem cells. PLoS One 2014; 9:e107596. [PMID: 25264898 PMCID: PMC4180068 DOI: 10.1371/journal.pone.0107596] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/05/2014] [Indexed: 12/18/2022] Open
Abstract
Currently, the treatment for ovarian cancer entails cytoreductive surgery followed by chemotherapy, mainly, carboplatin combined with paclitaxel. Although this regimen is initially effective in a high percentage of cases, unfortunately within few months of initial treatment, tumor relapse occurs because of platinum-resistance. This is attributed to chemo-resistance of cancer stem cells (CSCs). Herein we show for the first time that withaferin A (WFA), a bioactive compound isolated from the plant Withania somnifera, when used alone or in combination with cisplatin (CIS) targets putative CSCs. Treatment of nude mice bearing orthotopic ovarian tumors generated by injecting human ovarian epithelial cancer cell line (A2780) with WFA and cisplatin (WFA) alone or in combination resulted in a 70 to 80% reduction in tumor growth and complete inhibition of metastasis to other organs compared to untreated controls. Histochemical and Western blot analysis of the tumors revealed that inclusion of WFA (2 mg/kg) resulted in a highly significant elimination of cells expressing CSC markers - CD44, CD24, CD34, CD117 and Oct4 and downregulation of Notch1, Hes1 and Hey1 genes. In contrast treatment of mice with CIS alone (6 mg/kg) had opposite effect on those cells. Increase in cells expressing CSC markers and Notch1 signaling pathway in tumors exposed to CIS may explain recurrence of cancer in patients treated with carboplatin and paclitaxel. Since, WFA alone or in combination with CIS eliminates putative CSCs, we conclude that WFA in combination with CIS may present more efficacious therapy for ovarian cancer.
Collapse
Affiliation(s)
- Sham S. Kakar
- Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky, United States of America
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| | - Mariusz Z. Ratajczak
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Karen S. Powell
- Research Resources Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Mana Moghadamfalahi
- Department of Pathology, University of Louisville, Louisville, Kentucky, United States of America
| | - Donald M. Miller
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Sanjay K. Singh
- Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky, United States of America
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| |
Collapse
|
220
|
Yan B, Liu L, Zhao Y, Xiu LJ, Sun DZ, Liu X, Lu Y, Shi J, Zhang YC, Li YJ, Wang XW, Zhou YQ, Feng SH, Lv C, Wei PK, Qin ZF. Xiaotan Sanjie decoction attenuates tumor angiogenesis by manipulating Notch-1-regulated proliferation of gastric cancer stem-like cells. World J Gastroenterol 2014; 20:13105-13118. [PMID: 25278704 PMCID: PMC4177489 DOI: 10.3748/wjg.v20.i36.13105] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/10/2014] [Accepted: 05/19/2014] [Indexed: 02/06/2023] Open
Abstract
AIM To determine the underlying mechanisms of action and influence of Xiaotan Sanjie (XTSJ) decoction on gastric cancer stem-like cells (GCSCs). METHODS The gastric cancer cell line MKN-45 line was selected and sorted by FACS using the cancer stem cell marker CD44; the stemness of these cells was checked in our previous study. In an in vitro study, the expression of Notch-1, Hes1, Vascular endothelial growth factor (VEGF), and Ki-67 in both CD44-positive gastric cancer stem-like cells (GCSCs) and CD44-negative cells was measured by Western blot. The effect of XTSJ serum on cell viability and on the above markers was measured by MTT assay and Western blot, respectively. In an in vivo study, the ability to induce angiogenesis and maintenance of GCSCs in CD44-positive-MKN-45- and CD44-negative-engrafted mice were detected by immunohistochemical staining using markers for CD34 and CD44, respectively. The role of XTSJ decoction in regulating the expression of Notch-1, Hes1, VEGF and Ki-67 was measured by Western blot and real-time polymerase chain reaction. RESULTS CD44(+) GCSCs showed more cell proliferation and VEGF secretion than CD44-negative cells in vitro, which were accompanied by the high expression of Notch-1 and Hes1 and positively associated with tumor growth (GCSCs vs CD44-negative cells, 2.72 ± 0.25 vs 1.46 ± 0.16, P < 0.05) and microvessel density (MVD) (GCSCs vs CD44-negative cells, 8.15 ± 0.42 vs 3.83 ± 0.49, P < 0.001) in vivo. XTSJ decoction inhibited the viability of both cell types in a dose-dependent manner in vitro. Specifically, a significant difference in the medium- (82.87% ± 6.53%) and high-dose XTSJ groups (77.43% ± 7.34%) was detected at 24 h in the CD44(+) GCSCs group compared with the saline group (95.42% ± 5.76%) and the low-dose XTSJ group (90.74% ± 6.57%) (P < 0.05). However, the efficacy of XTSJ decoction was reduced in the CD44(-) groups; significant differences were only detected in the high-dose XTSJ group at 48 h (78.57% ± 6.94%) and 72 h (72.12% ± 7.68%) when compared with the other CD44- groups (P < 0.05). Notably, these differences were highly consistent with the Notch-1, Hes1, VEGF and Ki-67 expression in these cells. Similarly, in vivo, XTSJ decoction inhibited tumor growth in a dose-dependent manner. A significant difference was observed in the medium- (1.76 ± 0.15) and high-dose XTSJ (1.33 ± 0.081) groups compared with the GCSCs control group (2.72 ± 0.25) and the low-dose XTSJ group (2.51 ± 0.25) (P < 0.05). We also detected a remarkable decrease of MVD in the medium- (7.10 ± 0.60) and high-dose XTSJ (5.99 ± 0.47) groups compared with the GCSC control group (8.15 ± 0.42) and the low-dose XTSJ group (8.14 ± 0.46) (P < 0.05). Additionally, CD44 expression was decreased in these groups [medium- (4.43 ± 0.45) and high-dose XTSJ groups (3.56 ± 0.31) vs the GCSC control (5.96 ± 0.46) and low dose XTSJ groups (5.91 ± 0.38)] (P < 0.05). The significant differences in Notch-1, Hes1, VEGF and Ki-67 expression highly mirrored the results of XTSJ decoction in inhibiting tumor growth, MVD and CD44 expression. CONCLUSION Notch-1 may play an important role in regulating the proliferation of GCSCs; XTSJ decoction could attenuate tumor angiogenesis, at least partially, by inhibiting Notch-1.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Drugs, Chinese Herbal/pharmacology
- Gene Expression Regulation, Neoplastic
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Hyaluronan Receptors/genetics
- Hyaluronan Receptors/metabolism
- Ki-67 Antigen/genetics
- Ki-67 Antigen/metabolism
- Male
- Mice, Nude
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neovascularization, Pathologic
- Rats, Sprague-Dawley
- Receptor, Notch1/antagonists & inhibitors
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Signal Transduction/drug effects
- Stomach Neoplasms/blood supply
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Time Factors
- Transcription Factor HES-1
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Xenograft Model Antitumor Assays
Collapse
|
221
|
Radiosensitization by a novel Bcl-2 and Bcl-XL inhibitor S44563 in small-cell lung cancer. Cell Death Dis 2014; 5:e1423. [PMID: 25232677 PMCID: PMC4540189 DOI: 10.1038/cddis.2014.365] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/06/2014] [Accepted: 06/26/2014] [Indexed: 12/14/2022]
Abstract
Radiotherapy has a critical role in the treatment of small-cell lung cancer (SCLC). The effectiveness of radiation in SCLC remains limited as resistance results from defects in apoptosis. In the current study, we investigated whether using the Bcl-2/Bcl-XL inhibitor S44563 can enhance radiosensitivity of SCLC cells in vitro and in vivo. In vitro studies confirmed that S44563 caused SCLC cells to acquire hallmarks of apoptosis. S44563 markedly enhanced the sensitivity of SCLC cells to radiation, as determined by a clonogenic assay. The combination of S44563 and cisplatin-based chemo-radiation showed a significant tumor growth delay and increased overall survival in mouse xenograft models. This positive interaction was greater when S44563 was given after the completion of the radiation, which might be explained by the radiation-induced overexpression of anti-apoptotic proteins secondary to activation of the NF-κB pathway. These data underline the possibility of combining IR and Bcl-2/Bcl-XL inhibition in the treatment of SCLC as they underscore the importance of administering conventional and targeted therapies in an optimal sequence.
Collapse
|
222
|
Beaufort CM, Helmijr JCA, Piskorz AM, Hoogstraat M, Ruigrok-Ritstier K, Besselink N, Murtaza M, van IJcken WFJ, Heine AAJ, Smid M, Koudijs MJ, Brenton JD, Berns EMJJ, Helleman J. Ovarian cancer cell line panel (OCCP): clinical importance of in vitro morphological subtypes. PLoS One 2014; 9:e103988. [PMID: 25230021 PMCID: PMC4167545 DOI: 10.1371/journal.pone.0103988] [Citation(s) in RCA: 320] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 07/05/2014] [Indexed: 12/19/2022] Open
Abstract
Epithelial ovarian cancer is a highly heterogeneous disease and remains the most lethal gynaecological malignancy in the Western world. Therapeutic approaches need to account for inter-patient and intra-tumoural heterogeneity and detailed characterization of in vitro models representing the different histological and molecular ovarian cancer subtypes is critical to enable reliable preclinical testing. There are approximately 100 publicly available ovarian cancer cell lines but their cellular and molecular characteristics are largely undescribed. We have characterized 39 ovarian cancer cell lines under uniform conditions for growth characteristics, mRNA/microRNA expression, exon sequencing, drug response for clinically-relevant therapeutics and collated all available information on the original clinical features and site of origin. We tested for statistical associations between the cellular and molecular features of the lines and clinical features. Of the 39 ovarian cancer cell lines, 14 were assigned as high-grade serous, four serous-type, one low-grade serous and 20 non-serous type. Three morphological subtypes: Epithelial (n = 21), Round (n = 7) and Spindle (n = 12) were identified that showed distinct biological and molecular characteristics, including overexpression of cell movement and migration-associated genes in the Spindle subtype. Comparison with the original clinical data showed association of the spindle-like tumours with metastasis, advanced stage, suboptimal debulking and poor prognosis. In addition, the expression profiles of Spindle, Round and Epithelial morphologies clustered with the previously described C1-stromal, C5-mesenchymal and C4 ovarian subtype expression profiles respectively. Comprehensive profiling of 39 ovarian cancer cell lines under controlled, uniform conditions demonstrates clinically relevant cellular and genomic characteristics. This data provides a rational basis for selecting models to develop specific treatment approaches for histological and molecular subtypes of ovarian cancer.
Collapse
Affiliation(s)
- Corine M. Beaufort
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jean C. A. Helmijr
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Anna M. Piskorz
- Functional Genomics of Ovarian Cancer Laboratory, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Marlous Hoogstraat
- Department of Medical Oncology and Center for Personalized Cancer Treatment, University Medical Center Utrecht, Utrecht The Netherlands
| | | | - Nicolle Besselink
- Department of Medical Oncology and Center for Personalized Cancer Treatment, University Medical Center Utrecht, Utrecht The Netherlands
| | - Muhammed Murtaza
- Molecular and Computational Diagnostics Laboratory, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | | | - Anouk A. J. Heine
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marcel Smid
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marco J. Koudijs
- Department of Medical Oncology and Center for Personalized Cancer Treatment, University Medical Center Utrecht, Utrecht The Netherlands
| | - James D. Brenton
- Functional Genomics of Ovarian Cancer Laboratory, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Els M. J. J. Berns
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jozien Helleman
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
223
|
Meng E, Mitra A, Tripathi K, Finan MA, Scalici J, McClellan S, da Silva LM, Reed E, Shevde LA, Palle K, Rocconi RP. 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. [PMID: 25216266 PMCID: PMC4162571 DOI: 10.1371/journal.pone.0107142] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/07/2014] [Indexed: 12/17/2022] Open
Abstract
Objective Aldehyde dehydrogenase (ALDH) expressing cells have been characterized as possessing stem cell-like properties. We evaluated ALDH+ ovarian cancer stem cell-like properties and their role in platinum resistance. Methods Isogenic ovarian cancer cell lines for platinum sensitivity (A2780) and platinum resistant (A2780/CP70) as well as ascites from ovarian cancer patients were analyzed for ALDH+ by flow cytometry to determine its association to platinum resistance, recurrence and survival. A stable shRNA knockdown model for ALDH1A1 was utilized to determine its effect on cancer stem cell-like properties, cell cycle checkpoints, and DNA repair mediators. Results ALDH status directly correlated to platinum resistance in primary ovarian cancer samples obtained from ascites. Patients with ALDHHIGH displayed significantly lower progression free survival than the patients with ALDHLOW cells (9 vs. 3 months, respectively p<0.01). ALDH1A1-knockdown significantly attenuated clonogenic potential, PARP-1 protein levels, and reversed inherent platinum resistance. ALDH1A1-knockdown resulted in dramatic decrease of KLF4 and p21 protein levels thereby leading to S and G2 phase accumulation of cells. Increases in S and G2 cells demonstrated increased expression of replication stress associated Fanconi Anemia DNA repair proteins (FANCD2, FANCJ) and replication checkpoint (pS317 Chk1) were affected. ALDH1A1-knockdown induced DNA damage, evidenced by robust induction of γ-H2AX and BAX mediated apoptosis, with significant increases in BRCA1 expression, suggesting ALDH1A1-dependent regulation of cell cycle checkpoints and DNA repair networks in ovarian cancer stem-like cells. Conclusion This data suggests that ovarian cancer cells expressing ALDH1A1 may maintain platinum resistance by altered regulation of cell cycle checkpoint and DNA repair network signaling.
Collapse
Affiliation(s)
- Erhong Meng
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
| | - Aparna Mitra
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
| | - Kaushlendra Tripathi
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
| | - Michael A. Finan
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
| | - Jennifer Scalici
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
| | - Steve McClellan
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
| | - Luciana Madeira da Silva
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
| | - Eddie Reed
- National Institutes of Health, National Institute on Minority Health and Health Disparities, Bethesda, Maryland, United States of America
| | - Lalita A. Shevde
- University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Komaraiah Palle
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
- * E-mail: (KP); (RPR)
| | - Rodney P. Rocconi
- University of South Alabama Mitchell Cancer Institute, Mobile, Alabama, United States of America
- * E-mail: (KP); (RPR)
| |
Collapse
|
224
|
Anderson AS, Roberts PC, Frisard MI, Hulver MW, Schmelz EM. Ovarian tumor-initiating cells display a flexible metabolism. Exp Cell Res 2014; 328:44-57. [PMID: 25172556 DOI: 10.1016/j.yexcr.2014.08.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 08/14/2014] [Accepted: 08/17/2014] [Indexed: 01/06/2023]
Abstract
An altered metabolism during ovarian cancer progression allows for increased macromolecular synthesis and unrestrained growth. However, the metabolic phenotype of cancer stem or tumor-initiating cells, small tumor cell populations that are able to recapitulate the original tumor, has not been well characterized. In the present study, we compared the metabolic phenotype of the stem cell enriched cell variant, MOSE-LFFLv (TIC), derived from mouse ovarian surface epithelial (MOSE) cells, to their parental (MOSE-L) and benign precursor (MOSE-E) cells. TICs exhibit a decrease in glucose and fatty acid oxidation with a concomitant increase in lactate secretion. In contrast to MOSE-L cells, TICs can increase their rate of glycolysis to overcome the inhibition of ATP synthase by oligomycin and can increase their oxygen consumption rate to maintain proton motive force when uncoupled, similar to the benign MOSE-E cells. TICs have an increased survival rate under limiting conditions as well as an increased survival rate when treated with AICAR, but exhibit a higher sensitivity to metformin than MOSE-E and MOSE-L cells. Together, our data show that TICs have a distinct metabolic profile that may render them flexible to adapt to the specific conditions of their microenvironment. By better understanding their metabolic phenotype and external environmental conditions that support their survival, treatment interventions can be designed to extend current therapy regimens to eradicate TICs.
Collapse
Affiliation(s)
- Angela S Anderson
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Paul C Roberts
- Biomedical Science and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Madlyn I Frisard
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Matthew W Hulver
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA.
| | - Eva M Schmelz
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
225
|
Dreaden EC, Morton SW, Shopsowitz KE, Choi JH, Deng ZJ, Cho NJ, Hammond PT. Bimodal tumor-targeting from microenvironment responsive hyaluronan layer-by-layer (LbL) nanoparticles. ACS NANO 2014; 8:8374-82. [PMID: 25100313 PMCID: PMC4148172 DOI: 10.1021/nn502861t] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 08/06/2014] [Indexed: 05/21/2023]
Abstract
Active targeting of nanoscale drug carriers can improve tumor-specific delivery; however, cellular heterogeneity both within and among tumor sites is a fundamental barrier to their success. Here, we describe a tumor microenvironment-responsive layer-by-layer (LbL) polymer drug carrier that actively targets tumors based on two independent mechanisms: pH-dependent cellular uptake at hypoxic tumor pH and hyaluronan-directed targeting of cell-surface CD44 receptor, a well-characterized biomarker for breast and ovarian cancer stem cells. Hypoxic pH-induced structural reorganization of hyaluronan-LbL nanoparticles was a direct result of the nature of the LbL electrostatic complex, and led to targeted cellular delivery in vitro and in vivo, with effective tumor penetration and uptake. The nanoscale drug carriers selectively bound CD44 and diminished cancer cell migration in vitro, while co-localizing with the CD44 receptor in vivo. Multimodal targeting of LbL nanoparticles is a powerful strategy for tumor-specific cancer diagnostics and therapy that can be accomplished using a single bilayer of polyamine and hyaluronan that, when assembled, produce a dynamic and responsive cell-particle interface.
Collapse
Affiliation(s)
- Erik C. Dreaden
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Stephen W. Morton
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Kevin E. Shopsowitz
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Jae-Hyeok Choi
- School of Materials Science and Engineering, Centre for Biomimetic Sensor Science, and School of Chemical and Biomedical Engineering, Nanyang Technological University, 639798 Singapore
| | - Zhou J. Deng
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Centre for Biomimetic Sensor Science, and School of Chemical and Biomedical Engineering, Nanyang Technological University, 639798 Singapore
| | - Paula T. Hammond
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
- Address correspondence to
| |
Collapse
|
226
|
Yang-Hartwich Y, Gurrea-Soteras M, Sumi N, Joo WD, Holmberg JC, Craveiro V, Alvero AB, Mor G. Ovulation and extra-ovarian origin of ovarian cancer. Sci Rep 2014; 4:6116. [PMID: 25135607 PMCID: PMC4137344 DOI: 10.1038/srep06116] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/25/2014] [Indexed: 12/16/2022] Open
Abstract
The mortality rate of ovarian cancer remains high due to late diagnosis and recurrence. A fundamental step toward improving detection and treatment of this lethal disease is to understand its origin. A growing number of studies have revealed that ovarian cancer can develop from multiple extra-ovarian origins, including fallopian tube, gastrointestinal tract, cervix and endometriosis. However, the mechanism leading to their ovarian localization is not understood. We utilized in vitro, ex vivo, and in vivo models to recapitulate the process of extra-ovarian malignant cells migrating to the ovaries and forming tumors. We provided experimental evidence to support that ovulation, by disrupting the ovarian surface epithelium and releasing chemokines/cytokines, promotes the migration and adhesion of malignant cells to the ovary. We identified the granulosa cell-secreted SDF-1 as a main chemoattractant that recruits malignant cells towards the ovary. Our findings revealed a potential molecular mechanism of how the extra-ovarian cells can be attracted by the ovary, migrate to and form tumors in the ovary. Our data also supports the association between increased ovulation and the risk of ovarian cancer. Understanding this association will lead us to the development of more specific markers for early detection and better prevention strategies.
Collapse
Affiliation(s)
- Yang Yang-Hartwich
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marta Gurrea-Soteras
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Natalia Sumi
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Won Duk Joo
- CHA Bundang Medical Center, CHA University, South Korea
| | - Jennie C Holmberg
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Vinicius Craveiro
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ayesha B Alvero
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gil Mor
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
227
|
Reversing paclitaxel resistance in ovarian cancer cells via inhibition of the ABCB1 expressing side population. Tumour Biol 2014; 35:9879-92. [PMID: 24993095 DOI: 10.1007/s13277-014-2277-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/24/2014] [Indexed: 12/18/2022] Open
Abstract
The majority of deaths in ovarian cancer are caused by recurrent metastatic disease which is usually multidrug resistant. This progression has been hypothesised to be due in part to the presence of cancer stem cells, a subset of cells which are capable of self-renewal and are able to survive chemotherapy and migrate to distant sites. Side population (SP) cells, identified by the efflux of the DNA-binding dye Hoechst 33342 through ATP-binding cassette (ABC) transporters, are a known adult stem cell group and have been suggested as a cancer stem cell in various cancers. Despite the identification of SP cells in cancer cell lines and patient samples, little attention has been paid to the identification of specific ABC transporters within this cell fraction which efflux Hoechst dye and thus may facilitate drug resistance. In this study, we demonstrate that SP cells can be detected in both ovarian cancer cell lines and ascitic fluid samples, and these SP cells possess stem cell and drug resistance properties. We show that ABCB1 is the functioning ABC transporter in ovarian cancer cell lines, and expression of ABCB1 is associated with a paclitaxel-resistant phenotype. Moreover, silencing of ABCB1 using a specific morpholino oligonucleotide results in an inhibition of the SP phenotype and a sensitising of ovarian cancer cell lines to paclitaxel. ABCB1 should therefore be considered as a therapeutic target in ovarian cancer.
Collapse
|
228
|
Ectopic over-expression of miR-429 induces mesenchymal-to-epithelial transition (MET) and increased drug sensitivity in metastasizing ovarian cancer cells. Gynecol Oncol 2014; 134:96-103. [DOI: 10.1016/j.ygyno.2014.04.055] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/29/2014] [Indexed: 11/22/2022]
|
229
|
d'Adhemar CJ, Spillane CD, Gallagher MF, Bates M, Costello KM, Barry-O'Crowley J, Haley K, Kernan N, Murphy C, Smyth PC, O'Byrne K, Pennington S, Cooke AA, Ffrench B, Martin CM, O'Donnell D, Hennessy B, Stordal B, Finn S, McCann A, Gleeson N, D'Arcy T, Flood B, O'Neill LAJ, Sheils O, O'Toole S, O'Leary JJ. The MyD88+ phenotype is an adverse prognostic factor in epithelial ovarian cancer. PLoS One 2014; 9:e100816. [PMID: 24977712 PMCID: PMC4076208 DOI: 10.1371/journal.pone.0100816] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/30/2014] [Indexed: 11/18/2022] Open
Abstract
The prognosis of epithelial ovarian cancer is poor in part due to the high frequency of chemoresistance. Recent evidence points to the Toll-like receptor-4 (TLR4), and particularly its adaptor protein MyD88, as one potential mediator of this resistance. This study aims to provide further evidence that MyD88 positive cancer cells are clinically significant, stem-like and reproducibly detectable for the purposes of prognostic stratification. Expression of TLR4 and MyD88 was assessed immunohistochemically in 198 paraffin-embedded ovarian tissues and in an embryonal carcinoma model of cancer stemness. In parallel, expression of TLR4 and MyD88 mRNA and regulatory microRNAs (miR-21 and miR-146a) was assessed, as well as in a series of chemosensitive and resistant cancer cells lines. Functional analysis of the pathway was assessed in chemoresistant SKOV-3 ovarian cancer cells. TLR4 and MyD88 expression can be reproducibly assessed via immunohistochemistry using a semi-quantitative scoring system. TLR4 expression was present in all ovarian epithelium (normal and neoplastic), whereas MyD88 was restricted to neoplastic cells, independent of tumour grade and associated with reduced progression-free and overall survival, in an immunohistological specific subset of serous carcinomas, p<0.05. MiR-21 and miR-146a expression was significantly increased in MyD88 negative cancers (p<0.05), indicating their participation in regulation. Significant alterations in MyD88 mRNA expression were observed between chemosensitive and chemoresistant cells and tissue. Knockdown of TLR4 in SKOV-3 ovarian cells recovered chemosensitivity. Knockdown of MyD88 alone did not. MyD88 expression was down-regulated in differentiated embryonal carcinoma (NTera2) cells, supporting the MyD88+ cancer stem cell hypothesis. Our findings demonstrate that expression of MyD88 is associated with significantly reduced patient survival and altered microRNA levels and suggest an intact/functioning TLR4/MyD88 pathway is required for acquisition of the chemoresistant phenotype. Ex vivo manipulation of ovarian cancer stem cell (CSC) differentiation can decrease MyD88 expression, providing a potentially valuable CSC model for ovarian cancer.
Collapse
MESH Headings
- Aged
- Antineoplastic Agents, Phytogenic/pharmacology
- Carcinoma, Ovarian Epithelial
- Cell Line, Tumor
- Cystadenocarcinoma, Serous/diagnosis
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/mortality
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Genotype
- Humans
- Immunohistochemistry
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Myeloid Differentiation Factor 88/genetics
- Myeloid Differentiation Factor 88/metabolism
- Neoplasms, Glandular and Epithelial/diagnosis
- Neoplasms, Glandular and Epithelial/drug therapy
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/mortality
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Ovarian Neoplasms/diagnosis
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/mortality
- Paclitaxel/pharmacology
- Phenotype
- Prognosis
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Survival Analysis
- Toll-Like Receptor 4/antagonists & inhibitors
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
| | - Cathy D. Spillane
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | | | - Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Katie M. Costello
- Department of Pathology, Coombe Women's & Infants University Hospital, Dublin, Ireland
| | | | - Kathryn Haley
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - Niamh Kernan
- Department of Pathology, Coombe Women's & Infants University Hospital, Dublin, Ireland
| | - Ciara Murphy
- Department of Pathology, Coombe Women's & Infants University Hospital, Dublin, Ireland
| | - Paul C. Smyth
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Ken O'Byrne
- Translational Research Institute, Princess Alexandra Hospital, Brisbane, Australia
| | - Stephen Pennington
- College of Health Sciences, University College Dublin, Belfield, Dublin, Ireland
| | - Aoife A. Cooke
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Brendan Ffrench
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Cara M. Martin
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | | | - Bryan Hennessy
- Department of Medical Oncology, Royal College of Surgeons, Dublin, Ireland
| | - Britta Stordal
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Stephen Finn
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Amanda McCann
- College of Health Sciences, University College Dublin, Belfield, Dublin, Ireland
| | - Noreen Gleeson
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - Tom D'Arcy
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - Brian Flood
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | | | - Orla Sheils
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
- * E-mail:
| | - John J. O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Department of Pathology, Coombe Women's & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
230
|
Kim TH, Suh DH, Kim MK, Song YS. Metformin against cancer stem cells through the modulation of energy metabolism: special considerations on ovarian cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:132702. [PMID: 25050322 PMCID: PMC4094711 DOI: 10.1155/2014/132702] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/29/2014] [Indexed: 12/17/2022]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy among women worldwide and is presumed to result from the presence of ovarian cancer stem cells. To overcome the limitation of current anticancer agents, another anticancer strategy is necessary to effectively target cancer stem cells in ovarian cancer. In many types of malignancies, including ovarian cancer, metformin, one of the most popular antidiabetic drugs, has been demonstrated to exhibit chemopreventive and anticancer efficacy with respect to incidence and overall survival rates. Thus, the metabolic reprogramming of cancer and cancer stem cells driven by genetic alterations during carcinogenesis and cancer progression could be therapeutically targeted. In this review, the potential efficacy and anticancer mechanisms of metformin against ovarian cancer stem cells will be discussed.
Collapse
Affiliation(s)
- Tae Hun Kim
- Department of Obstetrics and Gynecology, Korean Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam 463-707, Republic of Korea
| | - Mi-Kyung Kim
- Biomedical Science Project, Brain Korea 21 Program for Leading Universities & Students, Seoul National University, Seoul 110-799, Republic of Korea
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
- Major in Biomodulation, World Class University, Seoul National University, Seoul 151-921, Republic of Korea
| |
Collapse
|
231
|
Hun Lee J, Shu L, Fuentes F, Su ZY, Tony Kong AN. Cancer chemoprevention by traditional chinese herbal medicine and dietary phytochemicals: targeting nrf2-mediated oxidative stress/anti-inflammatory responses, epigenetics, and cancer stem cells. J Tradit Complement Med 2014; 3:69-79. [PMID: 24716158 PMCID: PMC3924975 DOI: 10.4103/2225-4110.107700] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Excessive oxidative stress induced by reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive metabolites of carcinogens alters cellular homeostasis, leading to genetic/epigenetic changes, genomic instability, neoplastic transformation, and cancer initiation/progression. As a protective mechanism against oxidative stress, antioxidant/detoxifying enzymes reduce these reactive species and protect normal cells from endo-/exogenous oxidative damage. The transcription factor nuclear factor-erythroid 2 p45 (NF-E2)-related factor 2 (Nrf2), a master regulator of the antioxidative stress response, plays a critical role in the expression of many cytoprotective enzymes, including NAD(P)H:quinine oxidoreductase (NQO1), heme oxygenase-1 (HO-1), UDP-glucuronosyltransferase (UGT), and glutathione S-transferase (GST). Recent studies demonstrated that many dietary phytochemicals derived from various vegetables, fruits, spices, and herbal medicines induce Nrf2-mediated antioxidant/detoxifying enzymes, restore aberrant epigenetic alterations, and eliminate cancer stem cells (CSCs). The Nrf2-mediated antioxidant response prevents many age-related diseases, including cancer. Owing to their fundamental contribution to carcinogenesis, epigenetic modifications and CSCs are novel targets of dietary phytochemicals and traditional Chinese herbal medicine (TCHM). In this review, we summarize cancer chemoprevention by dietary phytochemicals, including TCHM, which have great potential as a safer and more effective strategy for preventing cancer.
Collapse
Affiliation(s)
- Jong Hun Lee
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Limin Shu
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Francisco Fuentes
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA ; Department of Desert Agriculture and Biotechnology, Arturo Prat University, PO box 121, Iquique, Chile
| | - Zheng-Yuan Su
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Ah-Ng Tony Kong
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| |
Collapse
|
232
|
Falci C, Dieci MV, Guarneri V, Soldà C, Bria E, Tortora G, Conte P. Maintenance therapy in epithelial ovarian cancer: from chemotherapy to targeted agents. Expert Rev Anticancer Ther 2014; 14:1041-50. [PMID: 24953376 DOI: 10.1586/14737140.2014.922415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Over the past years, although no increase in the cure rate for advanced epithelial ovarian cancer patients has been achieved, a slow prolongation in patients survival has been observed, thanks to the introduction of effective second line or salvage therapies. Attempts to disease chronicization seem therefore of value in this setting. A major effort has been pursued to establish the role of maintenance therapies for epithelial ovarian cancer patients. Although chemotherapy does not seem to have an effective role, promising results are coming from trials investigating maintenance targeted treatments, especially with antiangiogenic agents or PARP inhibitors for selected patients. The aim of this article is to review current evidences on maintenance therapy for epithelial ovarian cancer and put the results in perspective.
Collapse
Affiliation(s)
- Cristina Falci
- Division of Medical Oncology 2, Istituto Oncologico Veneto IRCCS, via Gattamelata 64, 35128, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
233
|
Condello S, Morgan CA, Nagdas S, Cao L, Turek J, Hurley TD, Matei D. β-Catenin-regulated ALDH1A1 is a target in ovarian cancer spheroids. Oncogene 2014; 34:2297-308. [PMID: 24954508 PMCID: PMC4275429 DOI: 10.1038/onc.2014.178] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 05/10/2014] [Accepted: 05/12/2014] [Indexed: 01/06/2023]
Abstract
Cancer cells form three dimensional (3D) multicellular aggregates (or
spheroids) under non-adherent culture conditions. In ovarian cancer (OC),
spheroids serve as a vehicle for cancer cell dissemination in the peritoneal
cavity, protecting cells from environmental stress-induced anoikis. To identify
new targetable molecules in OC spheroids, we investigated gene expression
profiles and networks upregulated in three dimensional (3D) versus traditional
monolayer culture conditions. We identified ALDH1A1, a cancer
stem cell marker as being overexpressed in OC spheroids and directly connected
to key elements of the β-catenin pathway. B-catenin function and
ALDH1A1 expression were increased in OC spheroids vs.
monolayers and in successive spheroid generations, suggesting that 3D aggregates
are enriched in cells with stem cell characteristics. B-catenin knockdown
decreased ALDH1A1 expression levels and β-catenin
coimmunoprecipitated with the ALDH1A1 promoter, suggesting that
ALDH1A1 is a direct β-catenin target. Both siRNA
mediated β-catenin knockdown and A37, a novel ALDH1A1 small molecule
enzymatic inhibitor described here for the first time, disrupted OC spheroid
formation and cell viability (p<0.001). B-catenin knockdown blocked tumor
growth and peritoneal metastasis in an OC xenograft model. These data strongly
support the role of β-catenin regulated ALDH1A1 in the maintenance of OC
spheroids and propose new ALDH1A1 inhibitors targeting this cell population.
Collapse
Affiliation(s)
- S Condello
- Department of Medicine, Indianapolis, IN, USA
| | - C A Morgan
- Department of Biochemistry and Molecular Biology, Indianapolis, IN, USA
| | - S Nagdas
- University of Virginia Medical School, Indianapolis, IN, USA
| | - L Cao
- Department of Medicine, Indianapolis, IN, USA
| | - J Turek
- College of Veterinary Medicine Purdue University, Indianapolis, IN, USA
| | - T D Hurley
- 1] Department of Biochemistry and Molecular Biology, Indianapolis, IN, USA [2] Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - D Matei
- 1] Department of Medicine, Indianapolis, IN, USA [2] Department of Biochemistry and Molecular Biology, Indianapolis, IN, USA [3] Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA [4] VA Roudebush Hospital, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
234
|
Colvin EK. Tumor-associated macrophages contribute to tumor progression in ovarian cancer. Front Oncol 2014; 4:137. [PMID: 24936477 PMCID: PMC4047518 DOI: 10.3389/fonc.2014.00137] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/21/2014] [Indexed: 01/21/2023] Open
Abstract
Ovarian cancer is the leading cause of death in women with gynecological malignancy and improvements in current treatments are needed. As with many other solid cancers, the ovarian tumor microenvironment is emerging as a key player in tumor progression and a potential therapeutic target. The tumor microenvironment contains several non-malignant cell types that are known to contribute to tumor progression and metastasis. Included in this population of non-malignant cells are several different types of immune cells, of which tumor-associated macrophages (TAMs) are the most abundant. An increasing amount of evidence is emerging to suggest that TAMs display a unique activation profile in ovarian tumors and are able to create an immunosuppressive microenvironment, allowing tumors to evade immune detection and promoting tumor progression. Therefore, an increased understanding of how these immune cells interact with tumor cells and the microenvironment will greatly benefit the development of more effective immunotherapies to treat ovarian cancer. This review focuses on the role of TAMs in the ovarian tumor microenvironment and how they promote tumor progression.
Collapse
Affiliation(s)
- Emily K Colvin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney , St. Leonards, NSW , Australia
| |
Collapse
|
235
|
Abubaker K, Luwor RB, Zhu H, McNally O, Quinn MA, Burns CJ, Thompson EW, Findlay JK, Ahmed N. Inhibition of the JAK2/STAT3 pathway in ovarian cancer results in the loss of cancer stem cell-like characteristics and a reduced tumor burden. BMC Cancer 2014; 14:317. [PMID: 24886434 PMCID: PMC4025194 DOI: 10.1186/1471-2407-14-317] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 04/23/2014] [Indexed: 12/21/2022] Open
Abstract
Background Current treatment of ovarian cancer patients with chemotherapy leaves behind a residual tumor which results in recurrent ovarian cancer within a short time frame. We have previously demonstrated that a single short-term treatment of ovarian cancer cells with chemotherapy in vitro resulted in a cancer stem cell (CSC)-like enriched residual population which generated significantly greater tumor burden compared to the tumor burden generated by control untreated cells. In this report we looked at the mechanisms of the enrichment of CSC-like residual cells in response to paclitaxel treatment. Methods The mechanism of survival of paclitaxel-treated residual cells at a growth inhibitory concentration of 50% (GI50) was determined on isolated tumor cells from the ascites of recurrent ovarian cancer patients and HEY ovarian cancer cell line by in vitro assays and in a mouse xenograft model. Results Treatment of isolated tumor cells from the ascites of ovarian cancer patients and HEY ovarian cancer cell line with paclitaxel resulted in a CSC-like residual population which coincided with the activation of Janus activated kinase 2 (JAK2) and signal transducer and activation of transcription 3 (STAT3) pathway in paclitaxel surviving cells. Both paclitaxel-induced JAK2/STAT3 activation and CSC-like characteristics were inhibited by a low dose JAK2-specific small molecule inhibitor CYT387 (1 μM) in vitro. Subsequent, in vivo transplantation of paclitaxel and CYT387-treated HEY cells in mice resulted in a significantly reduced tumor burden compared to that seen with paclitaxel only-treated transplanted cells. In vitro analysis of tumor xenografts at protein and mRNA levels demonstrated a loss of CSC-like markers and CA125 expression in paclitaxel and CYT387-treated cell-derived xenografts, compared to paclitaxel only-treated cell-derived xenografts. These results were consistent with significantly reduced activation of JAK2 and STAT3 in paclitaxel and CYT387-treated cell-derived xenografts compared to paclitaxel only-treated cell derived xenografts. Conclusions This proof of principle study demonstrates that inhibition of the JAK2/STAT3 pathway by the addition of CYT387 suppresses the ‘stemness’ profile in chemotherapy-treated residual cells in vitro, which is replicated in vivo, leading to a reduced tumor burden. These findings have important implications for ovarian cancer patients who are treated with taxane and/or platinum-based therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Nuzhat Ahmed
- Women's Cancer Research Centre, Royal Women's Hospital, 20 Flemington Road, Parkville, Melbourne, Victoria 3052, Australia.
| |
Collapse
|
236
|
Abubaker K, Luwor RB, Escalona R, McNally O, Quinn MA, Thompson EW, Findlay JK, Ahmed N. Targeted Disruption of the JAK2/STAT3 Pathway in Combination with Systemic Administration of Paclitaxel Inhibits the Priming of Ovarian Cancer Stem Cells Leading to a Reduced Tumor Burden. Front Oncol 2014; 4:75. [PMID: 24782986 PMCID: PMC3988380 DOI: 10.3389/fonc.2014.00075] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/25/2014] [Indexed: 12/15/2022] Open
Abstract
Chemotherapy resistance associated with recurrent disease is the major cause of poor survival of ovarian cancer patients. We have recently demonstrated activation of the JAK2/STAT3 pathway and the enhancement of a cancer stem cell (CSC)-like phenotype in ovarian cancer cells treated in vitro with chemotherapeutic agents. To elucidate further these mechanisms in vivo, we used a two-tiered paclitaxel treatment approach in nude mice inoculated with ovarian cancer cells. In the first approach, we demonstrate that a single intraperitoneal administration of paclitaxel in mice 7 days after subcutaneous transplantation of the HEY ovarian cancer cell line resulted in a significant increase in the expression of CA125, Oct4, and CD117 in mice xenografts compared to control mice xenografts which did not receive paclitaxel. In the second approach, mice were administered once weekly with paclitaxel and/or a daily dose of the JAK2-specific inhibitor, CYT387, over 4 weeks. Mice receiving paclitaxel only demonstrated a significant decrease in tumor volume compared to control mice. At the molecular level, mouse tumors remaining after paclitaxel administration showed a significant increase in the expression of Oct4 and CD117 coinciding with a significant activation of the JAK2/STAT3 pathway compared to control tumors. The addition of CYT387 with paclitaxel resulted in the suppression of JAK2/STAT3 activation and abrogation of Oct4 and CD117 expression in mouse xenografts. This coincided with significantly smaller tumors in mice administered CYT387 in addition to paclitaxel, compared to the control group and the group of mice receiving paclitaxel only. These data suggest that the systemic administration of paclitaxel enhances Oct4- and CD117-associated CSC-like marker expression in surviving cancer cells in vivo, which can be suppressed by the addition of the JAK2-specific inhibitor CYT387, leading to a significantly smaller tumor burden. These novel findings have the potential for the development of CSC-targeted therapy to improve the treatment outcomes of ovarian cancer patients.
Collapse
Affiliation(s)
- Khalid Abubaker
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Surgery, St Vincent Hospital, University of Melbourne , Melbourne, VIC , Australia
| | - Rodney B Luwor
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne , Melbourne, VIC , Australia
| | - Ruth Escalona
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia
| | - Orla McNally
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Melbourne, VIC , Australia
| | - Michael A Quinn
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Melbourne, VIC , Australia
| | - Erik W Thompson
- Department of Surgery, St Vincent Hospital, University of Melbourne , Melbourne, VIC , Australia ; St Vincent's Institute , Fitzroy, VIC , Australia
| | - Jock K Findlay
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Melbourne, VIC , Australia ; Prince Henry's Institute of Medical Research , Clayton, VIC , Australia
| | - Nuzhat Ahmed
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Surgery, St Vincent Hospital, University of Melbourne , Melbourne, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Melbourne, VIC , Australia ; Prince Henry's Institute of Medical Research , Clayton, VIC , Australia
| |
Collapse
|
237
|
Shetzer Y, Solomon H, Koifman G, Molchadsky A, Horesh S, Rotter V. The paradigm of mutant p53-expressing cancer stem cells and drug resistance. Carcinogenesis 2014; 35:1196-208. [PMID: 24658181 DOI: 10.1093/carcin/bgu073] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is well accepted that expression of mutant p53 involves the gain of oncogenic-specific activities accentuating the malignant phenotype. Depending on the specific cancer type, mutant p53 can contribute to either the early or the late events of the multiphase process underlying the transformation of a normal cell into a cancerous one. This multifactorial system is evident in ~50% of human cancers. Mutant p53 was shown to interfere with a variety of cellular functions that lead to augmented cell survival, cellular plasticity, aberration of DNA repair machinery and other effects. All these effects culminate in the acquisition of drug resistance often seen in cancer cells. Interestingly, drug resistance has also been suggested to be associated with cancer stem cells (CSCs), which reside within growing tumors. The notion that p53 plays a regulatory role in the life of stem cells, coupled with the observations that p53 mutations may contribute to the evolvement of CSCs makes it challenging to speculate that drug resistance and cancer recurrence are mediated by CSCs expressing mutant p53.
Collapse
Affiliation(s)
- Yoav Shetzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hilla Solomon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gabriela Koifman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alina Molchadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Stav Horesh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
238
|
Tomao F, Papa A, Strudel M, Rossi L, Lo Russo G, Benedetti Panici P, Ciabatta FR, Tomao S. Investigating molecular profiles of ovarian cancer: an update on cancer stem cells. J Cancer 2014; 5:301-10. [PMID: 24723972 PMCID: PMC3982176 DOI: 10.7150/jca.8610] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 02/09/2014] [Indexed: 12/14/2022] Open
Abstract
Currently we are more and more improving our knowledge about the characteristics and the role of cancer stem cells in human cancer. Particularly we have realized that self-renewing ovarian cancer stem cells (CSCs) or ovarian cancer-initiating cells, and mesenchymal stem cells (SCs) too, are probably implicated in the etiopathogenesis of epithelial ovarian cancer (EOC). There is clear evidence that these cells are also involved in its intra- and extra-peritoneal diffusion and in the occurrence of chemo-resistance. In assessing the molecular characteristics of ovarian CSCs, we have to take note that these cellular populations are rare and the absence of specific cell surface markers represents a challenge to isolate and identify pure SC populations. In our review, we focused our attention on the molecular characteristics of epithelial ovarian CSCs and on the methods to detect them starting from their biological features. The study of ovarian CSCs is taking on an increasingly important strategic role, mostly for the potential therapeutic application in the next future.
Collapse
Affiliation(s)
- Federica Tomao
- 1. Department of Gynecology and Obstetrics, Policlinico Umberto I Hospital, University of Rome, Italy
| | - Anselmo Papa
- 2. Oncology Unit, ICOT Hospital, Policlinico Umberto I Hospital, University of Rome, Italy
| | - Martina Strudel
- 2. Oncology Unit, ICOT Hospital, Policlinico Umberto I Hospital, University of Rome, Italy
| | - Luigi Rossi
- 2. Oncology Unit, ICOT Hospital, Policlinico Umberto I Hospital, University of Rome, Italy
| | - Giuseppe Lo Russo
- 2. Oncology Unit, ICOT Hospital, Policlinico Umberto I Hospital, University of Rome, Italy
| | | | | | - Silverio Tomao
- 2. Oncology Unit, ICOT Hospital, Policlinico Umberto I Hospital, University of Rome, Italy
| |
Collapse
|
239
|
Ribeiro JR, Lovasco LA, Vanderhyden BC, Freiman RN. Targeting TBP-Associated Factors in Ovarian Cancer. Front Oncol 2014; 4:45. [PMID: 24653979 PMCID: PMC3949196 DOI: 10.3389/fonc.2014.00045] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/25/2014] [Indexed: 12/11/2022] Open
Abstract
As ovarian tumors progress, they undergo a process of dedifferentiation, allowing adaptive changes in growth and morphology that promote metastasis and chemoresistance. Herein, we outline a hypothesis that TATA-box binding protein associated factors (TAFs), which compose the RNA Polymerase II initiation factor, TFIID, contribute to regulation of dedifferentiation states in ovarian cancer. Numerous studies demonstrate that TAFs regulate differentiation and proliferation states; their expression is typically high in pluripotent cells and reduced upon differentiation. Strikingly, TAF2 exhibits copy number increases or mRNA overexpression in 73% of high-grade serous ovarian cancers (HGSC). At the biochemical level, TAF2 directs TFIID to TATA-less promoters by contact with an Initiator element, which may lead to the deregulation of the transcriptional output of these tumor cells. TAF4, which is altered in 66% of HGSC, is crucial for the stability of the TFIID complex and helps drive dedifferentiation of mouse embryonic fibroblasts to induced pluripotent stem cells. Its ovary-enriched paralog, TAF4B, is altered in 26% of HGSC. Here, we show that TAF4B mRNA correlates with Cyclin D2 mRNA expression in human granulosa cell tumors. TAF4B may also contribute to regulation of tumor microenvironment due to its estrogen-responsiveness and ability to act as a cofactor for NFκB. Conversely, TAF9, a cofactor for p53 in regulating apoptosis, may act as a tumor suppressor in ovarian cancer, since it is downregulated or deleted in 98% of HGSC. We conclude that a greater understanding of mechanisms of transcriptional regulation that execute signals from oncogenic signaling cascades is needed in order to expand our understanding of the etiology and progression of ovarian cancer, and most importantly to identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
| | - Lindsay A Lovasco
- Molecular and Cellular Biology and Biochemistry, Brown University , Providence, RI , USA
| | - Barbara C Vanderhyden
- Cellular and Molecular Medicine, University of Ottawa , Ottawa, ON , Canada ; Centre for Cancer Therapeutics, Ottawa Hospital Research Institute , Ottawa, ON , Canada
| | - Richard N Freiman
- Pathobiology Graduate Program, Brown University , Providence, RI , USA ; Molecular and Cellular Biology and Biochemistry, Brown University , Providence, RI , USA
| |
Collapse
|
240
|
Londero AP, Orsaria M, Tell G, Marzinotto S, Capodicasa V, Poletto M, Vascotto C, Sacco C, Mariuzzi L. Expression and prognostic significance of APE1/Ref-1 and NPM1 proteins in high-grade ovarian serous cancer. Am J Clin Pathol 2014; 141:404-14. [PMID: 24515769 DOI: 10.1309/ajcpidkdlsge26cx] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES To correlate the expression profile of human apurinic endonuclease/redox factor 1 (APE1/Ref-1) with that of nucleolar/nucleoplasmic protein nucleophosmin 1 (NPM1) in association with the aggressiveness and progression of high-grade ovarian serous cancer. METHODS Retrospective study analyzing a tissue microarray of 73 women affected by high-grade ovarian serous cancer. Protein expression was assessed by immunohistochemistry on primary tumor masses and synchronous peritoneal metastases if present. RESULTS APE1/Ref-1 and NPM1 showed a significant correlation in ovarian serous cancer. Patients with a poorer outcome showed a significant overexpression of nuclear NPM1 protein. A Cox proportional hazards multivariate regression model revealed NPM1 expression to be independently significant for overall survival in high-grade ovarian serous cancers after correcting for stage, age, cytoreduction completeness, and platinum resistance. CONCLUSIONS APE1/Ref-1 interacts with NPM1 to control the DNA damage repair system, and it is likely that this interaction plays a defining role in high-grade ovarian serous carcinoma. A high NPM1 immunohistochemical expression was independently correlated with a shorter survival period and thus appears to be an important prognostic factor.
Collapse
Affiliation(s)
- Ambrogio P. Londero
- Deparment of Experimental Clinical and Medical Science, Clinic of Obstetrics and Gynecology, University of Udine, Udine, Italy
| | - Maria Orsaria
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Gianluca Tell
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Stefania Marzinotto
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Valentina Capodicasa
- Deparment of Experimental Clinical and Medical Science, Clinic of Obstetrics and Gynecology, University of Udine, Udine, Italy
| | - Mattia Poletto
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Carlo Vascotto
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Cosimo Sacco
- Clinic of Oncology, University Hospital of Udine, Udine, Italy
| | - Laura Mariuzzi
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| |
Collapse
|
241
|
Han C, Zhao R, Liu X, Srivastava A, Gong L, Mao H, Qu M, Zhao W, Yu J, Wang QE. DDB2 suppresses tumorigenicity by limiting the cancer stem cell population in ovarian cancer. Mol Cancer Res 2014; 12:784-94. [PMID: 24574518 DOI: 10.1158/1541-7786.mcr-13-0638] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
UNLABELLED Ovarian cancer is an extremely aggressive disease associated with a high percentage of tumor recurrence and chemotherapy resistance. Understanding the underlying mechanism of tumor relapse is crucial for effective therapy of ovarian cancer. DNA damage-binding protein 2 (DDB2) is a DNA repair factor mainly involved in nucleotide excision repair. Here, a novel role was identified for DDB2 in the tumorigenesis of ovarian cancer cells and the prognosis of patients with ovarian cancer. Overexpressing DDB2 in human ovarian cancer cells suppressed its capability to recapitulate tumors in athymic nude mice. Mechanistic investigation demonstrated that DDB2 is able to reduce the cancer stem cell (CSC) population characterized with high aldehyde dehydrogenase activity in ovarian cancer cells, probably through disrupting the self-renewal capacity of CSCs. Low DDB2 expression correlates with poor outcomes among patients with ovarian cancer, as revealed from the analysis of publicly available gene expression array datasets. Given the finding that DDB2 protein expression is low in ovarian tumor cells, enhancement of DDB2 expression is a promising strategy to eradicate CSCs and would help to halt ovarian cancer relapse. IMPLICATIONS DDB2 status has prognostic potential, and elevating its expression eradicates CSCs and could reduce ovarian cancer relapse.
Collapse
Affiliation(s)
- Chunhua Han
- Authors' Affiliations: Departments of Radiology and 2Pathology; 3Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio; and 4Weifang Medical University, Shandong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Chen Z, Forman LW, Williams RM, Faller DV. Protein kinase C-δ inactivation inhibits the proliferation and survival of cancer stem cells in culture and in vivo. BMC Cancer 2014; 14:90. [PMID: 24528676 PMCID: PMC3927586 DOI: 10.1186/1471-2407-14-90] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/06/2014] [Indexed: 12/18/2022] Open
Abstract
Background A subpopulation of tumor cells with distinct stem-like properties (cancer stem-like cells, CSCs) may be responsible for tumor initiation, invasive growth, and possibly dissemination to distant organ sites. CSCs exhibit a spectrum of biological, biochemical, and molecular features that are consistent with a stem-like phenotype, including growth as non-adherent spheres (clonogenic potential), ability to form a new tumor in xenograft assays, unlimited self-renewal, and the capacity for multipotency and lineage-specific differentiation. PKCδ is a novel class serine/threonine kinase of the PKC family, and functions in a number of cellular activities including cell proliferation, survival or apoptosis. PKCδ has previously been validated as a synthetic lethal target in cancer cells of multiple types with aberrant activation of Ras signaling, using both genetic (shRNA and dominant-negative PKCδ mutants) and small molecule inhibitors. In contrast, PKCδ is not required for the proliferation or survival of normal cells, suggesting the potential tumor-specificity of a PKCδ-targeted approach. Methods shRNA knockdown was used validate PKCδ as a target in primary cancer stem cell lines and stem-like cells derived from human tumor cell lines, including breast, pancreatic, prostate and melanoma tumor cells. Novel and potent small molecule PKCδ inhibitors were employed in assays monitoring apoptosis, proliferation and clonogenic capacity of these cancer stem-like populations. Significant differences among data sets were determined using two-tailed Student’s t tests or ANOVA. Results We demonstrate that CSC-like populations derived from multiple types of human primary tumors, from human cancer cell lines, and from transformed human cells, require PKCδ activity and are susceptible to agents which deplete PKCδ protein or activity. Inhibition of PKCδ by specific genetic strategies (shRNA) or by novel small molecule inhibitors is growth inhibitory and cytotoxic to multiple types of human CSCs in culture. PKCδ inhibition efficiently prevents tumor sphere outgrowth from tumor cell cultures, with exposure times as short as six hours. Small-molecule PKCδ inhibitors also inhibit human CSC growth in vivo in a mouse xenograft model. Conclusions These findings suggest that the novel PKC isozyme PKCδ may represent a new molecular target for cancer stem cell populations.
Collapse
Affiliation(s)
| | | | | | - Douglas V Faller
- Cancer Center, Boston University School of Medicine, K-712C, 72 E, Concord St,, Boston, MA 02118, USA.
| |
Collapse
|
243
|
House CD, Hernandez L, Annunziata CM. Recent technological advances in using mouse models to study ovarian cancer. Front Oncol 2014; 4:26. [PMID: 24592355 PMCID: PMC3923136 DOI: 10.3389/fonc.2014.00026] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 01/28/2014] [Indexed: 12/14/2022] Open
Abstract
Serous epithelial ovarian cancer (SEOC) is the most lethal gynecological cancer in the United States with disease recurrence being the major cause of morbidity and mortality. Despite recent advances in our understanding of the molecular mechanisms responsible for the development of SEOC, the survival rate for women with this disease has remained relatively unchanged in the last two decades. Preclinical mouse models of ovarian cancer, including xenograft, syngeneic, and genetically engineered mice, have been developed to provide a mechanism for studying the development and progression of SEOC. Such models strive to increase our understanding of the etiology and dissemination of ovarian cancer in order to overcome barriers to early detection and resistance to standard chemotherapy. Although there is not a single model that is most suitable for studying ovarian cancer, improvements have led to current models that more closely mimic human disease in their genotype and phenotype. Other advances in the field, such as live animal imaging techniques, allow effective monitoring of the microenvironment and therapeutic efficacy. New and improved preclinical mouse models, combined with technological advances to study such models, will undoubtedly render success of future human clinical trials for patients with SEOC.
Collapse
Affiliation(s)
| | - Lidia Hernandez
- Women's Malignancies Branch, National Cancer Institute , Bethesda, MD , USA
| | | |
Collapse
|
244
|
Mimeault M, Batra SK. Molecular biomarkers of cancer stem/progenitor cells associated with progression, metastases, and treatment resistance of aggressive cancers. Cancer Epidemiol Biomarkers Prev 2014; 23:234-54. [PMID: 24273063 PMCID: PMC3977531 DOI: 10.1158/1055-9965.epi-13-0785] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The validation of novel diagnostic, prognostic, and predictive biomarkers and therapeutic targets in tumor cells is of critical importance for optimizing the choice and efficacy of personalized therapies. Importantly, recent advances have led to the identification of gene-expression signatures in cancer cells, including cancer stem/progenitor cells, in the primary tumors, exosomes, circulating tumor cells (CTC), and disseminated cancer cells at distant metastatic sites. The gene-expression signatures may help to improve the accuracy of diagnosis and predict the therapeutic responses and overall survival of patients with cancer. Potential biomarkers in cancer cells include stem cell-like markers [CD133, aldehyde dehydrogenase (ALDH), CD44, and CD24], growth factors, and their cognate receptors [epidermal growth factor receptor (EGFR), EGFRvIII, and HER2], molecules associated with epithelial-mesenchymal transition (EMT; vimentin, N-cadherin, snail, twist, and Zeb1), regulators of altered metabolism (phosphatidylinositol-3' kinase/Akt/mTOR), and drug resistance (multidrug transporters and macrophage inhibitory cytokine-1). Moreover, different pluripotency-associated transcription factors (Oct3/4, Nanog, Sox2, and Myc) and microRNAs that are involved in the epigenetic reprogramming and acquisition of stem cell-like properties by cancer cells during cancer progression may also be exploited as molecular biomarkers to predict the risk of metastases, systemic treatment resistance, and disease relapse of patients with cancer.
Collapse
Affiliation(s)
- Murielle Mimeault
- Authors' Affiliation: Department of Biochemistry and Molecular Biology, Fred & Pamela Buffet Cancer Center, Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, Nebraska
| | | |
Collapse
|
245
|
Kalir T, Firpo-Betancourt A, Nezhat F. Update on ovarian cancer pathogenesis: history, controversies, emerging issues and future impact. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17474108.2013.847638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
246
|
Vaz AP, Ponnusamy MP, Seshacharyulu P, Batra SK. A concise review on the current understanding of pancreatic cancer stem cells. JOURNAL OF CANCER STEM CELL RESEARCH 2014; 2:e1004. [PMID: 26451384 PMCID: PMC4594952 DOI: 10.14343/jcscr.2014.2e1004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Several evidences suggest that a small population of cells known as cancer stem cells (CSCs) or tumor initiating stemlike cells within a tumor is capable of tumor initiation, maintenance and propagation. Recent publications have supported the existence of CSCs in pancreatic tumors. The pancreatic stem/progenitor cells, which express self-renewal markers, are identified to be present in the peribiliary gland. Based on the CSC hypothesis, mutations can lead to the transformation of stem/progenitor cells or differentiated cells into CSCs. The pancreatic CSCs express a wide array of markers such as CD44, CD24, ESA, CD133, c-MET, CXCR4, PD2/Paf1 and ALDH1. The CSCs are isolated based on surface markers or by other methods such as ALDEFLOUR assay or Hoechst 33342 dye exclusion assay. The isolated cells are further characterized by in vitro and in vivo tumorigenic assays. The most important characteristics of CSCs are its ability to self-renew and impart drug resistance towards chemotherapy. Moreover, these distinct cells display alteration of signaling pathways pertaining to CSCs such as Notch, Wnt and Shh to maintain the self-renewal process. Failure of cancer treatment could be attributed to the therapy resistance exhibited by the CSCs. Metastasis and drug resistance in pancreatic cancer is associated with epithelial to mesenchymal transition (EMT). Furthermore, mucins, the high molecular weight proteins are found to be associated with pancreatic CSCs and EMT. Understanding the underlying molecular pathways that aid in the metastatic and drug resistant nature of these distinct cells will aid in targeting these cells. Overall, this review focuses on the various aspects of pancreatic adult/stem progenitors, CSC hypothesis, its markers, pathways, niche, EMT and novel therapeutic drugs used for the elimination of pancreatic CSCs.
Collapse
Affiliation(s)
- Arokia Priyanka Vaz
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P. Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute for Research in Cancer and Allied Diseases and Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
247
|
Shah MM, Landen CN. Ovarian cancer stem cells: are they real and why are they important? Gynecol Oncol 2013; 132:483-9. [PMID: 24321398 DOI: 10.1016/j.ygyno.2013.12.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/25/2013] [Accepted: 12/02/2013] [Indexed: 12/15/2022]
Abstract
The cancer stem cell hypothesis has been put forward as a paradigm to describe varying levels of aggressiveness in heterogeneous tumors. Specifically, many subpopulations have been clearly demonstrated to possess increased tumorigenicity in mice, broad differentiating capacity, and resistance to therapy. However, the extent to which these experimental findings are potentially clinically significant is still not clear. This review will describe the principles of this emerging hypothesis, ways in which it may be appropriate in ovarian cancer based on the clinical course of the disease, and how we might exploit it to improve outcomes in ovarian cancer patients.
Collapse
Affiliation(s)
- Monjri M Shah
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles N Landen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
248
|
Joo WD, Visintin I, Mor G. Targeted cancer therapy--are the days of systemic chemotherapy numbered? Maturitas 2013; 76:308-14. [PMID: 24128673 PMCID: PMC4610026 DOI: 10.1016/j.maturitas.2013.09.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022]
Abstract
Targeted therapy or molecular targeted therapy has been defined as a type of treatment that blocks the growth of cancer cells by interfering with specific cell molecules required for carcinogenesis and tumor growth, rather than by simply interfering with all rapidly dividing cells as with traditional chemotherapy. There is a growing number of FDA approved monoclonal antibodies and small molecules targeting specific types of cancer suggestive of the growing relevance of this therapeutic approach. Targeted cancer therapies, also referred to as "Personalized Medicine", are being studied for use alone, in combination with other targeted therapies, and in combination with chemotherapy. The objective of personalized medicine is the identification of patients that would benefit from a specific treatment based on the expression of molecular markers. Examples of this approach include bevacizumab and olaparib, which have been designated as promising targeted therapies for ovarian cancer. Combinations of trastuzumab with pertuzumab, or T-DM1 and mTOR inhibitors added to an aromatase inhibitor are new therapeutic strategies for breast cancer. Although this approach has been seen as a major step in the expansion of personalized medicine, it has substantial limitations including its high cost and the presence of serious adverse effects. The Cancer Genome Atlas is a useful resource to identify novel and more effective targets, which may help to overcome the present limitations. In this review we will discuss the clinical outcome of some of these new therapies with a focus on ovarian and breast cancer. We will also discuss novel concepts in targeted therapy, the target of cancer stem cells.
Collapse
Affiliation(s)
- Won Duk Joo
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center, CHA University, Seoul, Republic of Korea
| | - Irene Visintin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Gil Mor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
249
|
Ahmed N, Stenvers KL. Getting to know ovarian cancer ascites: opportunities for targeted therapy-based translational research. Front Oncol 2013; 3:256. [PMID: 24093089 PMCID: PMC3782691 DOI: 10.3389/fonc.2013.00256] [Citation(s) in RCA: 335] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 09/11/2013] [Indexed: 01/13/2023] Open
Abstract
More than one third of ovarian cancer patients present with ascites at diagnosis, and almost all have ascites at recurrence. The presence of ascites correlates with the peritoneal spread of ovarian cancer and is associated with poor disease prognosis. Malignant ascites acts as a reservoir of a complex mixture of soluble factors and cellular components which provide a pro-inflammatory and tumor-promoting microenvironment for the tumor cells. Subpopulations of these tumor cells exhibit cancer stem-like phenotypes, possess enhanced resistance to therapies and the capacity for distal metastatic spread and recurrent disease. Thus, ascites-derived malignant cells and the ascites microenvironment represent a major source of morbidity and mortality for ovarian cancer patients. This review focuses on recent advances in our understanding of the molecular, cellular, and functional characteristics of the cellular populations within ascites and discusses their contributions to ovarian cancer metastasis, chemoresistance, and recurrence. We highlight in particular recent translational findings which have used primary ascites-derived tumor cells as a tool to understand the pathogenesis of the disease, yielding new insights and targets for therapeutic manipulation.
Collapse
Affiliation(s)
- Nuzhat Ahmed
- Women's Cancer Research Centre, Royal Women's Hospital , Parkville, VIC , Australia ; Department of Obstetrics and Gynaecology, University of Melbourne , Parkville, VIC , Australia ; Reproductive Development and Cancer Laboratory, Prince Henry's Institute for Medical Research , Melbourne, VIC , Australia
| | | |
Collapse
|
250
|
Fan YL, Zheng M, Tang YL, Liang XH. A new perspective of vasculogenic mimicry: EMT and cancer stem cells (Review). Oncol Lett 2013; 6:1174-1180. [PMID: 24179490 PMCID: PMC3813799 DOI: 10.3892/ol.2013.1555] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 07/17/2013] [Indexed: 02/05/2023] Open
Abstract
Vasculogenic mimicry (VM), a new pattern of tumor microcirculation, is important for the growth and progression of tumors. Epithelial-mesenchymal transition (EMT) is pivotal in malignant tumor progression and VM formation. With increasing knowledge of cancer stem cell (CSC) phenotypes and functions, increasing evidence suggests that CSCs are involved in VM formation. Recent studies have indicated that EMT is relevant to the acquisition and maintenance of stem cell-like characteristics. Thus, in this review we discuss the correlation between CSCs, EMT and VM formation.
Collapse
Affiliation(s)
- Yun-Long Fan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | | | | | | |
Collapse
|