1
|
Geoffroy K, Viens M, Kalin EM, Boudhraa Z, Roy DG, Wu JH, Provencher D, Mes-Masson AM, Bourgeois-Daigneault MC. The Small GTPase Ran Increases Sensitivity of Ovarian Cancer Cells to Oncolytic Vesicular Stomatitis Virus. Pharmaceuticals (Basel) 2024; 17:1662. [PMID: 39770503 PMCID: PMC11677601 DOI: 10.3390/ph17121662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Ovarian cancer is the deadliest gynecologic cancer, and with the majority of patients dying within the first five years of diagnosis, new therapeutic options are required. The small guanosine triphosphatase (GTPase) Ras-related nuclear protein (Ran) has been reported to be highly expressed in high-grade serous ovarian cancers (HGSOCs) and associated with poor outcomes. Blocking Ran function or preventing its expression were shown to be promising treatment strategies, however, there are currently no small molecule inhibitors available to specifically inhibit Ran function. Interestingly, a previous study suggested that the Vesicular stomatitis virus (VSV) could inhibit Ran activity. Given that VSV is an oncolytic virus (OV) and, therefore, has anti-cancer activity, we reasoned that oncolytic VSV (oVSV) might be particularly effective against ovarian cancer via Ran inhibition. Methods: We evaluated the sensitivity of patient-derived ovarian cancer cell lines to oVSV, as well as the impact of oVSV on Ran and vice versa, using overexpression systems, small interfering RNAs (siRNAs), and drug inhibition. Results: In this study, we evaluated the interplay between oVSV and Ran and found that, although oVSV does not consistently block Ran, increased Ran activation allows for better oVSV replication and tumor cell killing. Conclusions: Our study reveals a positive impact of Ran on oVSV sensitivity. Given the high expression of Ran in HGSOCs, which are particularly aggressive ovarian cancers, our data suggest that oVSV could be effective against the deadliest form of the disease.
Collapse
Affiliation(s)
- Karen Geoffroy
- Cancer Axis, Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada (M.V.); (E.M.K.); (D.G.R.); (D.P.)
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Mélissa Viens
- Cancer Axis, Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada (M.V.); (E.M.K.); (D.G.R.); (D.P.)
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Emma Mary Kalin
- Cancer Axis, Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada (M.V.); (E.M.K.); (D.G.R.); (D.P.)
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Zied Boudhraa
- Cancer Axis, Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada (M.V.); (E.M.K.); (D.G.R.); (D.P.)
- Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Dominic Guy Roy
- Cancer Axis, Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada (M.V.); (E.M.K.); (D.G.R.); (D.P.)
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Jian Hui Wu
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada;
| | - Diane Provencher
- Cancer Axis, Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada (M.V.); (E.M.K.); (D.G.R.); (D.P.)
- Division of Gynecologic Oncology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Anne-Marie Mes-Masson
- Cancer Axis, Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada (M.V.); (E.M.K.); (D.G.R.); (D.P.)
- Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Cancer Axis, Centre de Recherche du Centre Hospitalier de l’Université de Montréal and Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada (M.V.); (E.M.K.); (D.G.R.); (D.P.)
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
2
|
Geoffroy K, Mullins-Dansereau V, Leclerc-Desaulniers K, Viens M, Bourgeois-Daigneault MC. Oncolytic vesicular stomatitis virus alone or in combination with JAK inhibitors is effective against ovarian cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200826. [PMID: 39006945 PMCID: PMC11246050 DOI: 10.1016/j.omton.2024.200826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 07/16/2024]
Abstract
Therapy-resistant ovarian cancers have a poor prognosis and novel effective treatment options are urgently needed. In this study, we evaluated the therapeutic efficacy of the oncolytic vesicular stomatitis virus (VSV) against a panel of patient-derived ovarian cancer cell lines of all epithelial subtypes. Notably, we found that most of the cell lines were sensitive to VSV virotherapy. With the objective of improving treatment efficacy for the oncolytic virus-resistant cell lines, we tested various combinations with ovarian cancer standard of care drugs: olaparib, carboplatin, paclitaxel, doxorubicin, cyclophosphamide, and gemcitabine. While none of these combinations revealed to be beneficial, further experiments demonstrated that the antiviral interferon pathway was functional in VSV-resistant cell lines. Given that interferons signal through Janus kinase (JAK)-STAT to mediate their antiviral function, we tested combinations of oncolytic VSV with clinically relevant JAK inhibitors. Our results show that combining VSV with various JAK inhibitors, including ruxolitinib, enhances VSV virotherapy and treatment efficacy. Altogether, we show that VSV, either as a stand-alone treatment or in combination with JAK inhibitors provides an effective therapeutic option for ovarian cancer patients.
Collapse
Affiliation(s)
- Karen Geoffroy
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Victor Mullins-Dansereau
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Kim Leclerc-Desaulniers
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Mélissa Viens
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Cancer and Immunopathology Axes, CHUM Research Centre, Montreal, QC H2X 0A9, Canada
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
3
|
Mes-Masson AM. The journey from bench to bedside-it takes a science village. Biochem Cell Biol 2024; 102:299-304. [PMID: 38640502 DOI: 10.1139/bcb-2024-0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024] Open
Abstract
I was fortunate enough to start my career at what was the dawn of modern-day molecular biology and to apply it to an important health problem. While my early work focused on fundamental science, the desire to understand human disease better and to find practical applications for research discoveries resulted, over the following decades, in creating a stream of translational research directed specifically toward epithelial cancers. This could only have been possible through multiple collaborations. This type of team science would eventually become a hallmark of my career. With the development of higher throughput molecular techniques, the pace of research and discovery has quickened, and the concept of personalized medicine based on genomics is now coming to fruition. I hope my legacy will not just reflect my published works, but will also include the impact I have had on the development of the next generation of scientists and clinician scientists who inspired me with their dedication, knowledge, and enthusiasm.
Collapse
Affiliation(s)
- Anne-Marie Mes-Masson
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
4
|
Linh NT, Hang NT, Cuong BK, Linh DT, Phuong Linh NT, Nguyen-Van D, Dzung TN, Mao CV, Chung DT, Chinh LT, Hung NP, Tong HV, Toan NL. Establishment of cancer cell line originating from a patient with high-grade serous ovarian carcinoma. Future Sci OA 2023; 9:FSO875. [PMID: 37621847 PMCID: PMC10445588 DOI: 10.2144/fsoa-2023-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/22/2023] [Indexed: 08/26/2023] Open
Abstract
Aim Ovarian cancer is a serious malignancy with high prevalence and mortality. Methods We isolated and characterized an ovarian high-grade serous cancer cell line (M4) from a tumor of a Vietnamese patient with ovarian carcinoma. Results The M4 cancer cell line showed good proliferation and stability in culture. Morphologically, the M4 cells showed similar characteristics to tumor cells such as a polyhedral shape, large irregular nuclei, high nuclear/cytoplasmic ratio, high nuclear density and expressing cancer markers like CA125, p53 and Ki67 markers. Conclusion We have successfully isolated and characterized the M4 cell line from a Vietnamese patient with ovarian carcinoma.
Collapse
Affiliation(s)
- Nguyen Thuy Linh
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
- Department of Pathology, Military Hospital 103, Vietnam Military Medical University, Hanoi, Vietnam
- Department of Pathology, Hanoi Medical University Hanoi, Vietnam
| | - Ngo Thu Hang
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Bui Khac Cuong
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
| | - Dang Thuy Linh
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nham Thi Phuong Linh
- Laboratory Animal Research Center, Vietnam Military Medical University, Hanoi, Vietnam
| | - Do Nguyen-Van
- Department of Pathophysiology & Immunology, Hanoi Medical University, Hanoi, Vietnam
| | - Tran Ngoc Dzung
- Department of Pathology, Military Hospital 103, Vietnam Military Medical University, Hanoi, Vietnam
| | - Can Van Mao
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Dang Thanh Chung
- Department of Pathology, Military Hospital 103, Vietnam Military Medical University, Hanoi, Vietnam
| | - Le Tri Chinh
- Department of Gynaecology Surgery, Vietnam National Cancer Hospital, Hanoi, Vietnam
| | - Nguyen Phu Hung
- Faculty of Biotechnology, Thai Nguyen University of Sciences, Thai Nguyen, Vietnam
| | - Hoang Van Tong
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
- Institute of Biomedicine & Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nguyen Linh Toan
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| |
Collapse
|
5
|
Asare-Werehene M, Hunter RA, Gerber E, Reunov A, Brine I, Chang CY, Chang CC, Shieh DB, Burger D, Anis H, Tsang BK. The Application of an Extracellular Vesicle-Based Biosensor in Early Diagnosis and Prediction of Chemoresponsiveness in Ovarian Cancer. Cancers (Basel) 2023; 15:cancers15092566. [PMID: 37174032 PMCID: PMC10177169 DOI: 10.3390/cancers15092566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Ovarian cancer (OVCA) is the most fatal gynecological cancer with late diagnosis and plasma gelsolin (pGSN)-mediated chemoresistance representing the main obstacles to treatment success. Since there is no reliable approach to diagnosing patients at an early stage as well as predicting chemoresponsiveness, there is an urgent need to develop a diagnostic platform for such purposes. Small extracellular vesicles (sEVs) are attractive biomarkers given their potential accuracy for targeting tumor sites. METHODS We have developed a novel biosensor which utilizes cysteine-functionalized gold nanoparticles that simultaneously bind to cisplatin (CDDP) and plasma/cell-derived EVs, affording us the advantage of predicting OVCA chemoresponsiveness, and early diagnosis using surface-enhanced Raman spectroscopy. RESULTS We found that pGSN regulates cortactin (CTTN) content resulting in the formation of nuclear- and cytoplasmic-dense granules facilitating the secretion of sEVs carrying CDDP; a strategy used by resistant cells to survive CDDP action. The clinical utility of the biosensor was tested and subsequently revealed that the sEV/CA125 ratio outperformed CA125 and sEV individually in predicting early stage, chemoresistance, residual disease, tumor recurrence, and patient survival. CONCLUSION These findings highlight pGSN as a potential therapeutic target and provide a potential diagnostic platform to detect OVCA earlier and predict chemoresistance; an intervention that will positively impact patient-survival outcomes.
Collapse
Affiliation(s)
- Meshach Asare-Werehene
- Departments of Obstetrics & Gynecology and Cellular & Molecular Medicine, Centre for Infection, Immunity and Inflammation, Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
| | - Robert A Hunter
- School of Electrical Engineering and Computer Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Ottawa-Carleton Institute for Biomedical Engineering, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Emma Gerber
- Departments of Obstetrics & Gynecology and Cellular & Molecular Medicine, Centre for Infection, Immunity and Inflammation, Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
| | - Arkadiy Reunov
- Department of Biology, St. Francis Xavier University, 2320 Notre Dame Avenue, Antigonish, NS B2G 2W5, Canada
| | - Isaiah Brine
- Ottawa-Carleton Institute for Biomedical Engineering, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Chia-Yu Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Department of Electrophysics, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Chia-Ching Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan
- Department of Electrophysics, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
- Institute of Physics, Academia Sinica, Taipei 10529, Taiwan
| | - Dar-Bin Shieh
- Institute of Basic Medical Science, Institute of Oral Medicine and Department of Stomatology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan
- Advanced Optoelectronic Technology Center and Center for Micro/Nano Science and Technology, National Cheng Kung University, Tainan 701, Taiwan
| | - Dylan Burger
- Chronic Disease Program, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
| | - Hanan Anis
- School of Electrical Engineering and Computer Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Benjamin K Tsang
- Departments of Obstetrics & Gynecology and Cellular & Molecular Medicine, Centre for Infection, Immunity and Inflammation, Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
| |
Collapse
|
6
|
Sauriol SA, Carmona E, Udaskin ML, Radulovich N, Leclerc-Desaulniers K, Rottapel R, Oza AM, Lheureux S, Provencher DM, Mes-Masson AM. Inhibition of nicotinamide dinucleotide salvage pathway counters acquired and intrinsic poly(ADP-ribose) polymerase inhibitor resistance in high-grade serous ovarian cancer. Sci Rep 2023; 13:3334. [PMID: 36849518 PMCID: PMC9970983 DOI: 10.1038/s41598-023-30081-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023] Open
Abstract
Epithelial ovarian cancer is the most lethal gynecological malignancy, owing notably to its high rate of therapy-resistant recurrence in spite of good initial response to chemotherapy. Although poly(ADP-ribose) polymerase inhibitors (PARPi) have shown promise for ovarian cancer treatment, extended therapy usually leads to acquired PARPi resistance. Here we explored a novel therapeutic option to counter this phenomenon, combining PARPi and inhibitors of nicotinamide phosphoribosyltransferase (NAMPT). Cell-based models of acquired PARPi resistance were created through an in vitro selection procedure. Using resistant cells, xenograft tumors were grown in immunodeficient mice, while organoid models were generated from primary patient tumor samples. Intrinsically PARPi-resistant cell lines were also selected for analysis. Our results show that treatment with NAMPT inhibitors effectively sensitized all in vitro models to PARPi. Adding nicotinamide mononucleotide, the resulting NAMPT metabolite, abrogated the therapy-induced cell growth inhibition, demonstrating the specificity of the synergy. Treatment with olaparib (PARPi) and daporinad (NAMPT inhibitor) depleted intracellular NAD+ , induced double-strand DNA breaks, and promoted apoptosis as monitored by caspase-3 cleavage. The two drugs were also synergistic in mouse xenograft models and clinically relevant patient-derived organoids. Therefore, in the context of PARPi resistance, NAMPT inhibition could offer a promising new option for ovarian cancer patients.
Collapse
Affiliation(s)
- Skye Alexandre Sauriol
- Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, QC, H2X 0A9, Canada
- Institut du Cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Euridice Carmona
- Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, QC, H2X 0A9, Canada
- Institut du Cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Molly L Udaskin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Nikolina Radulovich
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Kim Leclerc-Desaulniers
- Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, QC, H2X 0A9, Canada
- Institut du Cancer de Montréal, Montreal, QC, H2X 0A9, Canada
| | - Robert Rottapel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Amit M Oza
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
- Division of Medical Oncology and Hematology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Stephanie Lheureux
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
- Division of Medical Oncology and Hematology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Diane M Provencher
- Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, QC, H2X 0A9, Canada
- Institut du Cancer de Montréal, Montreal, QC, H2X 0A9, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, QC, H2X 0A9, Canada.
- Institut du Cancer de Montréal, Montreal, QC, H2X 0A9, Canada.
- Department of Medicine, Université de Montréal, Montreal, QC, H3T 1J4, Canada.
| |
Collapse
|
7
|
Sevinyan L, Gupta P, Velliou E, Madhuri TK. The Development of a Three-Dimensional Platform for Patient-Derived Ovarian Cancer Tissue Models: A Systematic Literature Review. Cancers (Basel) 2022; 14:5628. [PMID: 36428724 PMCID: PMC9688222 DOI: 10.3390/cancers14225628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
There is an unmet biomedical need for ex vivo tumour models that would predict drug responses and in turn help determine treatment regimens and potentially predict resistance before clinical studies. Research has shown that three dimensional models of ovarian cancer (OvCa) are more realistic than two dimensional in vitro systems as they are able to capture patient in vivo conditions in more accurate manner. The vast majority of studies aiming to recapitulate the ovarian tumour morphology, behaviors, and study chemotherapy responses have been using ovarian cancer cell lines. However, despite the advantages of utilising cancer cell lines to set up a platform, they are not as informative as systems applying patient derived cells, as cell lines are not able to recapitulate differences between each individual patient characteristics. In this review we discussed the most recent advances in the creation of 3D ovarian cancer models that have used patient derived material, the challenges to overcome and future applications.
Collapse
Affiliation(s)
- Lusine Sevinyan
- Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
- Cancer Research, School of Applied Sciences, University of Brighton, Brighton BN2 4HQ, UK
| | - Priyanka Gupta
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK
| | - Thumuluru Kavitha Madhuri
- Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
- Cancer Research, School of Applied Sciences, University of Brighton, Brighton BN2 4HQ, UK
| |
Collapse
|
8
|
Caffeic acid phenethyl ester targets ubiquitin-specific protease 8 and synergizes with cisplatin in endometrioid ovarian carcinoma cells. Biochem Pharmacol 2022; 197:114900. [PMID: 34995485 DOI: 10.1016/j.bcp.2021.114900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 01/03/2023]
Abstract
Deubiquitinases (DUBs) mediate the removal of ubiquitin from diverse proteins that participate in the regulation of cell survival, DNA damage repair, apoptosis and drug resistance. Previous studies have shown an association between activation of cell survival pathways and platinum-drug resistance in ovarian carcinoma cell lines. Among the strategies available to inhibit DUBs, curcumin derivatives appear promising, thus we hypothesized their use to enhance the efficacy of cisplatin in ovarian carcinoma preclinical models. The caffeic acid phenethyl ester (CAPE), inhibited ubiquitin-specific protease 8 (USP8), but not proteasomal DUBs in cell-free assays. When CAPE was combined with cisplatin in nine cell lines representative of various histotypes a synergistic effect was observed in TOV112D cells and in the cisplatin-resistant IGROV-1/Pt1 variant, both of endometrioid type and carrying mutant TP53. In the latter cells, persistent G1 accumulation upon combined treatment associated with p27kip1 protein levels was observed. The synergy was not dependent on apoptosis induction, and appeared to occur in cells with higher USP8 levels. In vivo antitumor activity studies supported the advantage of the combination of CAPE and cisplatin in the subcutaneous model of cisplatin-resistant IGROV-1/Pt1 ovarian carcinoma as well as CAPE activity on intraperitoneal disease. This study reveals the therapeutic potential of CAPE in cisplatin-resistant ovarian tumors as well as in tumors expressing USP8.
Collapse
|
9
|
Boudhraa Z, Zaoui K, Fleury H, Cahuzac M, Gilbert S, Tchakarska G, Kendall-Dupont J, Carmona E, Provencher D, Mes-Masson AM. NR1D1 regulation by Ran GTPase via miR4472 identifies an essential vulnerability linked to aneuploidy in ovarian cancer. Oncogene 2021; 41:309-320. [PMID: 34743206 PMCID: PMC8755527 DOI: 10.1038/s41388-021-02082-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
While aneuploidy is a main enabling characteristic of cancers, it also creates specific vulnerabilities. Here we demonstrate that Ran inhibition targets epithelial ovarian cancer (EOC) survival through its characteristic aneuploidy. We show that induction of aneuploidy in rare diploid EOC cell lines or normal cells renders them highly dependent on Ran. We also establish an inverse correlation between Ran and the tumor suppressor NR1D1 and reveal the critical role of Ran/NR1D1 axis in aneuploidy-associated endogenous DNA damage repair. Mechanistically, we show that Ran, through the maturation of miR4472, destabilizes the mRNA of NR1D1 impacting several DNA repair pathways. We showed that NR1D1 interacts with both PARP1 and BRCA1 leading to the inhibition of DNA repair. Concordantly, loss of Ran was associated with NR1D1 induction, accumulation of DNA damages, and lethality of aneuploid EOC cells. Our findings suggest a synthetic lethal strategy targeting aneuploid cells based on their dependency to Ran.
Collapse
Affiliation(s)
- Zied Boudhraa
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada
| | - Kossay Zaoui
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada
| | - Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada
| | - Maxime Cahuzac
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada
| | - Sophie Gilbert
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada
| | - Guergana Tchakarska
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada
| | - Jennifer Kendall-Dupont
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada
| | - Euridice Carmona
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du cancer de Montréal (ICM), Montreal, QC, Canada.,Division of Gynecologic Oncology, Université de Montréal, Montreal, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada. .,Institut du cancer de Montréal (ICM), Montreal, QC, Canada. .,Department of Medicine, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
10
|
Carboplatin response in preclinical models for ovarian cancer: comparison of 2D monolayers, spheroids, ex vivo tumors and in vivo models. Sci Rep 2021; 11:18183. [PMID: 34521878 PMCID: PMC8440566 DOI: 10.1038/s41598-021-97434-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/04/2021] [Indexed: 12/18/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer. Among the key challenges in developing effective therapeutics is the poor translation of preclinical models used in the drug discovery pipeline. This leaves drug attrition rates and costs at an unacceptably high level. Previous work has highlighted the discrepancies in therapeutic response between current in vitro and in vivo models. To address this, we conducted a comparison study to differentiate the carboplatin chemotherapy response across four different model systems including 2D monolayers, 3D spheroids, 3D ex vivo tumors and mouse xenograft models. We used six previously characterized EOC cell lines of varying chemosensitivity and performed viability assays for each model. In vivo results from the mouse model correlated with 2D response in 3/6 cell lines while they correlated with 3D spheroids and the ex vivo model in 4/6 and 5/5 cell lines, respectively. Our results emphasize the variability in therapeutic response across models and demonstrate that the carboplatin response in EOC cell lines cultured in a 3D ex vivo model correlates best with the in vivo response. These results highlight a more feasible, reliable, and cost-effective preclinical model with the highest translational potential for drug screening and prediction studies in EOC.
Collapse
|
11
|
A Keratin 7 and E-Cadherin Signature Is Highly Predictive of Tubo-Ovarian High-Grade Serous Carcinoma Prognosis. Int J Mol Sci 2021; 22:ijms22105325. [PMID: 34070214 PMCID: PMC8158692 DOI: 10.3390/ijms22105325] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 01/06/2023] Open
Abstract
During tubo-ovarian high-grade serous carcinoma (HGSC) progression, tumoral cells undergo phenotypic changes in their epithelial marker profiles, which are essential for dissemination processes. Here, we set out to determine whether standard epithelial markers can predict HGSC patient prognosis. Levels of E-CADH, KRT7, KRT18, KRT19 were quantified in 18 HGSC cell lines by Western blot and in a Discovery cohort tissue microarray (TMA) (n = 101 patients) using immunofluorescence. E-CADH and KRT7 levels were subsequently analyzed in the TMA of the Canadian Ovarian Experimental Unified Resource cohort (COEUR, n = 1158 patients) and in public datasets. Epithelial marker expression was highly variable in HGSC cell lines and tissues. In the Discovery cohort, high levels of KRT7 and KRT19 were associated with an unfavorable prognosis, whereas high E-CADH expression indicated a better outcome. Expression of KRT7 and E-CADH gave a robust combination to predict overall survival (OS, p = 0.004) and progression free survival (PFS, p = 5.5 × 10−4) by Kaplan–Meier analysis. In the COEUR cohort, the E-CADH-KRT7 signature was a strong independent prognostic biomarker (OS, HR = 1.6, p = 2.9 × 10−4; PFS, HR = 1.3, p = 0.008) and predicted a poor patient response to chemotherapy (p = 1.3 × 10−4). Our results identify a combination of two epithelial markers as highly significant indicators of HGSC patient prognosis and treatment response.
Collapse
|
12
|
Amuzu S, Carmona E, Mes-Masson AM, Greenwood CMT, Tonin PN, Ragoussis J. Candidate Markers of Olaparib Response from Genomic Data Analyses of Human Cancer Cell Lines. Cancers (Basel) 2021; 13:1296. [PMID: 33803939 PMCID: PMC7998846 DOI: 10.3390/cancers13061296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
The benefit of PARP inhibitor olaparib in relapsed and advanced high-grade serous ovarian carcinoma (HGSOC) is well established especially in BRCA1/2 mutation carriers. Identification of additional biomarkers can help expand the population of patients most likely to benefit from olaparib treatment. To identify candidate markers of olaparib response we analyzed genomic and in vitro olaparib response data from two independent groups of cancer cell lines. Using pan-cancer cell lines (n = 896) from the Genomics of Drug Sensitivity in Cancer database, we applied linear regression methods to identify statistically significant gene predictors of olaparib response based on mRNA expression. We then analyzed whole exome sequencing and mRNA gene expression data from our collection of 18 HGSOC cell lines previously classified as sensitive, intermediate, or resistant based on in vitro olaparib response for mutations, copy number variation and differential expression of candidate olaparib response genes. We identify genes previously associated with olaparib response (SLFN11, ABCB1), and discover novel candidate olaparib sensitivity genes with known functions including interaction with PARP1 (PUM3, EEF1A1) and involvement in homologous recombination DNA repair (ELP4). Further investigations at experimental and clinical levels are required to validate novel candidates, and ultimately determine their efficacy as potential biomarkers of olaparib sensitivity.
Collapse
Affiliation(s)
- Setor Amuzu
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; (C.M.T.G.); (P.N.T.); (J.R.)
- McGill Genome Centre, McGill University, Montreal, QC H3A 0G1, Canada
| | - Euridice Carmona
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (E.C.); (A.-M.M.-M.)
- Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (E.C.); (A.-M.M.-M.)
- Institut du Cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Celia M. T. Greenwood
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; (C.M.T.G.); (P.N.T.); (J.R.)
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Departments of Oncology and Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC H3A 1A2, Canada
| | - Patricia N. Tonin
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; (C.M.T.G.); (P.N.T.); (J.R.)
- Cancer Research Program, Centre for Translational Biology, The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Department of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; (C.M.T.G.); (P.N.T.); (J.R.)
- McGill Genome Centre, McGill University, Montreal, QC H3A 0G1, Canada
| |
Collapse
|
13
|
Karnezis AN, Chen SY, Chow C, Yang W, Hendricks WPD, Ramos P, Briones N, Mes-Masson AM, Bosse T, Gilks CB, Trent JM, Weissman B, Huntsman DG, Wang Y. Re-assigning the histologic identities of COV434 and TOV-112D ovarian cancer cell lines. Gynecol Oncol 2021; 160:568-578. [PMID: 33328126 PMCID: PMC10039450 DOI: 10.1016/j.ygyno.2020.12.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/05/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The development of effective cancer treatments depends on the availability of cell lines that faithfully recapitulate the cancer in question. This study definitively re-assigns the histologic identities of two ovarian cancer cell lines, COV434 (originally described as a granulosa cell tumour) and TOV-112D (originally described as grade 3 endometrioid carcinoma), both of which were recently suggested to represent small cell carcinoma of the ovary, hypercalcemic type (SCCOHT), based on their shared gene expression profiles and sensitivity to EZH2 inhibitors. METHODS For COV434 and TOV-112D, we re-reviewed the original pathology slides and obtained clinical follow-up on the patients, when available, and performed immunohistochemistry for SMARCA4, SMARCA2 and additional diagnostic markers on the original formalin-fixed, paraffin-embedded (FFPE) clinical material, when available. For COV434, we further performed whole exome sequencing and validated SMARCA4 mutations by Sanger sequencing. We studied the growth of the cell lines at baseline and upon re-expression of SMARCA4 in vitro for both cell lines and evaluated the serum calcium levels in vivo upon injection into immunodeficient mice for COV434 cells. RESULTS The available morphological, immunohistochemical, genetic, and clinical features indicate COV434 is derived from SCCOHT, and TOV-112D is a dedifferentiated carcinoma. Transplantation of COV434 into mice leads to increased serum calcium level. Re-expression of SMARCA4 in either COV434 and TOV-112D cells suppressed their growth dramatically. CONCLUSIONS COV434 represents a bona fide SCCOHT cell line. TOV-112D is a dedifferentiated ovarian carcinoma cell line.
Collapse
MESH Headings
- Animals
- Carcinoma, Ovarian Epithelial/diagnosis
- Carcinoma, Ovarian Epithelial/drug therapy
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/pathology
- Carcinoma, Small Cell/diagnosis
- Carcinoma, Small Cell/drug therapy
- Carcinoma, Small Cell/genetics
- Carcinoma, Small Cell/pathology
- Cell Dedifferentiation/genetics
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/pathology
- DNA Helicases/analysis
- DNA Helicases/deficiency
- DNA Helicases/genetics
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Female
- Gene Expression Profiling
- Humans
- Mice
- Nuclear Proteins/analysis
- Nuclear Proteins/deficiency
- Nuclear Proteins/genetics
- Ovarian Neoplasms/diagnosis
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- Transcription Factors/analysis
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Exome Sequencing
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Anthony N Karnezis
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Shary Yuting Chen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Christine Chow
- Genetic Pathology Evaluation Centre, Vancouver General Hospital and University of British Columbia, Vancouver, BC, Canada
| | - Winnie Yang
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - William P D Hendricks
- Division of Integrated Cancer Genomics, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Pilar Ramos
- Division of Integrated Cancer Genomics, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Natalia Briones
- Division of Integrated Cancer Genomics, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, QC, Canada; Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jeffrey M Trent
- Division of Integrated Cancer Genomics, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Bernard Weissman
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada; Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada.
| | - Yemin Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
14
|
Patra B, Lateef MA, Brodeur MN, Fleury H, Carmona E, Péant B, Provencher D, Mes-Masson AM, Gervais T. Carboplatin sensitivity in epithelial ovarian cancer cell lines: The impact of model systems. PLoS One 2021; 15:e0244549. [PMID: 33382759 PMCID: PMC7774933 DOI: 10.1371/journal.pone.0244549] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/13/2020] [Indexed: 12/26/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy in North America, underscoring the need for the development of new therapeutic strategies for the management of this disease. Although many drugs are pre-clinically tested every year, only a few are selected to be evaluated in clinical trials, and only a small number of these are successfully incorporated into standard care. Inaccuracies with the initial in vitro drug testing may be responsible for some of these failures. Drug testing is often performed using 2D monolayer cultures or 3D spheroid models. Here, we investigate the impact that these different in vitro models have on the carboplatin response of four EOC cell lines, and in particular how different 3D models (polydimethylsiloxane-based microfluidic chips and ultra low attachment plates) influence drug sensitivity within the same cell line. Our results show that carboplatin responses were observed in both the 3D spheroid models tested using apoptosis/cell death markers by flow cytometry. Contrary to previously reported observations, these were not associated with a significant decrease in spheroid size. For the majority of the EOC cell lines (3 out of 4) a similar carboplatin response was observed when comparing both spheroid methods. Interestingly, two cell lines classified as resistant to carboplatin in 2D cultures became sensitive in the 3D models, and one sensitive cell line in 2D culture showed resistance in 3D spheroids. Our results highlight the challenges of choosing the appropriate pre-clinical models for drug testing.
Collapse
Affiliation(s)
- Bishnubrata Patra
- Department of Engineering Physics and Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Muhammad Abdul Lateef
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Melica Nourmoussavi Brodeur
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Hubert Fleury
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Euridice Carmona
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Benjamin Péant
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
| | - Diane Provencher
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montréal, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
- * E-mail: (TG); (AMMM)
| | - Thomas Gervais
- Department of Engineering Physics and Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM) and Institut du Cancer de Montréal, Montréal, QC, Canada
- * E-mail: (TG); (AMMM)
| |
Collapse
|
15
|
Sauriol A, Simeone K, Portelance L, Meunier L, Leclerc-Desaulniers K, de Ladurantaye M, Chergui M, Kendall-Dupont J, Rahimi K, Carmona E, Provencher DM, Mes-Masson AM. Modeling the Diversity of Epithelial Ovarian Cancer through Ten Novel Well Characterized Cell Lines Covering Multiple Subtypes of the Disease. Cancers (Basel) 2020; 12:E2222. [PMID: 32784519 PMCID: PMC7465288 DOI: 10.3390/cancers12082222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/30/2022] Open
Abstract
Cancer cell lines are amongst the most important pre-clinical models. In the context of epithelial ovarian cancer, a highly heterogeneous disease with diverse subtypes, it is paramount to study a wide panel of models in order to draw a representative picture of the disease. As this lethal gynaecological malignancy has seen little improvement in overall survival in the last decade, it is all the more pressing to support future research with robust and diverse study models. Here, we describe ten novel spontaneously immortalized patient-derived ovarian cancer cell lines, detailing their respective mutational profiles and gene/biomarker expression patterns, as well as their in vitro and in vivo growth characteristics. Eight of the cell lines were classified as high-grade serous, while two were determined to be of the rarer mucinous and clear cell subtypes, respectively. Each of the ten cell lines presents a panel of characteristics reflective of diverse clinically relevant phenomena, including chemotherapeutic resistance, metastatic potential, and subtype-associated mutations and gene/protein expression profiles. Importantly, four cell lines formed subcutaneous tumors in mice, a key characteristic for pre-clinical drug testing. Our work thus contributes significantly to the available models for the study of ovarian cancer, supplying additional tools to better understand this complex disease.
Collapse
Affiliation(s)
- Alexandre Sauriol
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Kayla Simeone
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Lise Portelance
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Liliane Meunier
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Kim Leclerc-Desaulniers
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Manon de Ladurantaye
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Meriem Chergui
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Jennifer Kendall-Dupont
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Kurosh Rahimi
- Department of Pathology, Centre hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 3E4, Canada;
| | - Euridice Carmona
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
| | - Diane M. Provencher
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (A.S.); (K.S.); (L.P.); (L.M.); (K.L.-D.); (M.d.L.); (M.C.); (J.K.-D.); (E.C.); (D.M.P.)
- Institut du cancer de Montréal, Montreal, QC H2X 0A9, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
16
|
Kumar J, Kaur G, Ren R, Lu Y, Lin K, Li J, Huang Y, Patel A, Barton MC, Macfarlan T, Zhang X, Cheng X. KRAB domain of ZFP568 disrupts TRIM28-mediated abnormal interactions in cancer cells. NAR Cancer 2020; 2:zcaa007. [PMID: 32743551 PMCID: PMC7380489 DOI: 10.1093/narcan/zcaa007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/25/2020] [Accepted: 04/30/2020] [Indexed: 01/31/2023] Open
Abstract
Interactions of KRAB (Krüppel-associated box)-associated protein KAP1 [also known as TRIM28 (tripartite motif containing protein 28)] with DNA-binding KRAB zinc finger (KRAB-ZF) proteins silence many transposable elements during embryogenesis. However, in some cancers, TRIM28 is upregulated and interacts with different partners, many of which are transcription regulators such as EZH2 in MCF7 cells, to form abnormal repressive or activating complexes that lead to misregulation of genes. We ask whether a KRAB domain-the TRIM28 interaction domain present in native binding partners of TRIM28 that mediate repression of transposable elements-could be used as a tool molecule to disrupt aberrant TRIM28 complexes. Expression of KRAB domain containing fragments from a KRAB-ZF protein (ZFP568) in MCF7 cells, without the DNA-binding zinc fingers, inhibited TRIM28-EZH2 interactions and caused degradation of both TRIM28 and EZH2 proteins as well as other components of the EZH2-associated polycomb repressor 2 complex. In consequence, the product of EZH2 enzymatic activity, trimethylation of histone H3 lysine 27 level, was significantly reduced. The expression of a synthetic KRAB domain significantly inhibits the growth of breast cancer cells (MCF7) but has no effect on normal (immortalized) human mammary epithelial cells (MCF10a). Further, we found that TRIM28 is a positive regulator of TRIM24 protein levels, as observed previously in prostate cancer cells, and expression of the KRAB domain also lowered TRIM24 protein. Importantly, reduction of TRIM24 levels, by treatment with either the KRAB domain or a small-molecule degrader targeted to TRIM24, is accompanied by an elevated level of tumor suppressor p53. Taken together, this study reveals a novel mechanism for a TRIM28-associated protein stability network and establishes TRIM28 as a potential therapeutic target in cancers where TRIM28 is elevated. Finally, we discuss a potential mechanism of KRAB-ZF gene expression controlled by a regulatory feedback loop of TRIM28-KRAB.
Collapse
Affiliation(s)
- Janani Kumar
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gundeep Kaur
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ren Ren
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jia Li
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Yun Huang
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Anamika Patel
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Michelle C Barton
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Todd Macfarlan
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Xing Zhang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaodong Cheng
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
17
|
Ran promotes membrane targeting and stabilization of RhoA to orchestrate ovarian cancer cell invasion. Nat Commun 2019; 10:2666. [PMID: 31209254 PMCID: PMC6573066 DOI: 10.1038/s41467-019-10570-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 05/15/2019] [Indexed: 12/22/2022] Open
Abstract
Ran is a nucleocytoplasmic shuttle protein that is involved in cell cycle regulation, nuclear-cytoplasmic transport, and cell transformation. Ran plays an important role in cancer cell survival and cancer progression. Here, we show that, in addition to the nucleocytoplasmic localization of Ran, this GTPase is specifically associated with the plasma membrane/ruffles of ovarian cancer cells. Ran depletion has a drastic effect on RhoA stability and inhibits RhoA localization to the plasma membrane/ruffles and RhoA activity. We further demonstrate that the DEDDDL domain of Ran is required for the interaction with serine 188 of RhoA, which prevents RhoA degradation by the proteasome pathway. Moreover, the knockdown of Ran leads to a reduction of ovarian cancer cell invasion by impairing RhoA signalling. Our findings provide advanced insights into the mode of action of the Ran-RhoA signalling axis and may represent a potential therapeutic avenue for drug development to prevent ovarian tumour metastasis. Ran, a nucleus-cytoplasm shuttle protein, is implicated in cancer development and survival. Here, the authors show that Ran binds RhoA to impair its degradation and allow its localisation to the plasma membrane of ovarian cancer cells for tumour invasion.
Collapse
|
18
|
Fleury H, Malaquin N, Tu V, Gilbert S, Martinez A, Olivier MA, Sauriol A, Communal L, Leclerc-Desaulniers K, Carmona E, Provencher D, Mes-Masson AM, Rodier F. Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat Commun 2019. [PMID: 31186408 DOI: 10.1038/s41467-019-10460-1] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Senescence is a tumor suppression mechanism defined by stable proliferation arrest. Here we demonstrate that the known synthetic lethal interaction between poly(ADP-ribose) polymerase 1 inhibitors (PARPi) and DNA repair triggers p53-independent ovarian cancer cell senescence defined by senescence-associated phenotypic hallmarks including DNA-SCARS, inflammatory secretome, Bcl-XL-mediated apoptosis resistance, and proliferation restriction via Chk2 and p21 (CDKN1A). The concept of senescence as irreversible remains controversial and here we show that PARPi-senescent cells re-initiate proliferation upon drug withdrawal, potentially explaining the requirement for sustained PARPi therapy in the clinic. Importantly, PARPi-induced senescence renders ovarian and breast cancer cells transiently susceptible to second-phase synthetic lethal approaches targeting the senescence state using senolytic drugs. The combination of PARPi and a senolytic is effective in preclinical models of ovarian and breast cancer suggesting that coupling these synthetic lethalities provides a rational approach to their clinical use and may together be more effective in limiting resistance.
Collapse
Affiliation(s)
- Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Véronique Tu
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Sophie Gilbert
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Aurélie Martinez
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Marc-Alexandre Olivier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Alexandre Sauriol
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Laudine Communal
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Kim Leclerc-Desaulniers
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Euridice Carmona
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada.,Division of Gynecologic Oncology, Université de Montréal, Montreal, H3C 3J7, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada. .,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada. .,Department of Medicine, Université de Montréal, Montreal, H3C 3J7, QC, Canada.
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada. .,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada. .,Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, H3C 3J7, QC, Canada.
| |
Collapse
|
19
|
Fleury H, Malaquin N, Tu V, Gilbert S, Martinez A, Olivier MA, Sauriol SA, Communal L, Leclerc-Desaulniers K, Carmona E, Provencher D, Mes-Masson AM, Rodier F. Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat Commun 2019; 10:2556. [PMID: 31186408 PMCID: PMC6560032 DOI: 10.1038/s41467-019-10460-1] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/09/2019] [Indexed: 12/19/2022] Open
Abstract
Senescence is a tumor suppression mechanism defined by stable proliferation arrest. Here we demonstrate that the known synthetic lethal interaction between poly(ADP-ribose) polymerase 1 inhibitors (PARPi) and DNA repair triggers p53-independent ovarian cancer cell senescence defined by senescence-associated phenotypic hallmarks including DNA-SCARS, inflammatory secretome, Bcl-XL-mediated apoptosis resistance, and proliferation restriction via Chk2 and p21 (CDKN1A). The concept of senescence as irreversible remains controversial and here we show that PARPi-senescent cells re-initiate proliferation upon drug withdrawal, potentially explaining the requirement for sustained PARPi therapy in the clinic. Importantly, PARPi-induced senescence renders ovarian and breast cancer cells transiently susceptible to second-phase synthetic lethal approaches targeting the senescence state using senolytic drugs. The combination of PARPi and a senolytic is effective in preclinical models of ovarian and breast cancer suggesting that coupling these synthetic lethalities provides a rational approach to their clinical use and may together be more effective in limiting resistance.
Collapse
Affiliation(s)
- Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Véronique Tu
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Sophie Gilbert
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Aurélie Martinez
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Marc-Alexandre Olivier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Skye Alexandre Sauriol
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Laudine Communal
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Kim Leclerc-Desaulniers
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Euridice Carmona
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montreal, H3C 3J7, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada.
- Department of Medicine, Université de Montréal, Montreal, H3C 3J7, QC, Canada.
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada.
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, H3C 3J7, QC, Canada.
| |
Collapse
|
20
|
Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat Commun 2019. [PMID: 31186408 DOI: 10.1038/s41467-019-10460-1]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Senescence is a tumor suppression mechanism defined by stable proliferation arrest. Here we demonstrate that the known synthetic lethal interaction between poly(ADP-ribose) polymerase 1 inhibitors (PARPi) and DNA repair triggers p53-independent ovarian cancer cell senescence defined by senescence-associated phenotypic hallmarks including DNA-SCARS, inflammatory secretome, Bcl-XL-mediated apoptosis resistance, and proliferation restriction via Chk2 and p21 (CDKN1A). The concept of senescence as irreversible remains controversial and here we show that PARPi-senescent cells re-initiate proliferation upon drug withdrawal, potentially explaining the requirement for sustained PARPi therapy in the clinic. Importantly, PARPi-induced senescence renders ovarian and breast cancer cells transiently susceptible to second-phase synthetic lethal approaches targeting the senescence state using senolytic drugs. The combination of PARPi and a senolytic is effective in preclinical models of ovarian and breast cancer suggesting that coupling these synthetic lethalities provides a rational approach to their clinical use and may together be more effective in limiting resistance.
Collapse
|
21
|
St-Georges-Robillard A, Cahuzac M, Péant B, Fleury H, Lateef MA, Ricard A, Sauriol A, Leblond F, Mes-Masson AM, Gervais T. Long-term fluorescence hyperspectral imaging of on-chip treated co-culture tumour spheroids to follow clonal evolution. Integr Biol (Camb) 2019; 11:130-141. [PMID: 31172192 DOI: 10.1093/intbio/zyz012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/09/2019] [Accepted: 05/30/2019] [Indexed: 12/22/2022]
Abstract
Multicellular tumour spheroids are an ideal in vitro tumour model to study clonal heterogeneity and drug resistance in cancer research because different cell types can be mixed at will. However, measuring the individual response of each cell population over time is challenging: current methods are either destructive, such as flow cytometry, or cannot image throughout a spheroid, such as confocal microscopy. Our group previously developed a wide-field fluorescence hyperspectral imaging system to study spheroids formed and cultured in microfluidic chips. In the present study, two subclones of a single parental ovarian cancer cell line transfected to express different fluorophores were produced and co-culture spheroids were formed on-chip using ratios forming highly asymmetric subpopulations. We performed a 3D proliferation assay on each cell population forming the spheroids that matched the 2D growth behaviour. Response assays to PARP inhibitors and platinum-based drugs were also performed to follow the clonal evolution of mixed populations. Our experiments show that hyperspectral imaging can detect spheroid response before observing a decrease in spheroid diameter. Hyperspectral imaging and microfluidic-based spheroid assays provide a versatile solution to study clonal heterogeneity, able to measure response in subpopulations presenting as little as 10% of the initial spheroid.
Collapse
Affiliation(s)
- Amélie St-Georges-Robillard
- Polytechnique Montréal, Department of Engineering Physics and Institute of Biomedical Engineering, Montreal, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Maxime Cahuzac
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Benjamin Péant
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
- TransMedTech Institute, Montréal, Canada
| | - Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Muhammad Abdul Lateef
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Alexis Ricard
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Alexandre Sauriol
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Frédéric Leblond
- Polytechnique Montréal, Department of Engineering Physics and Institute of Biomedical Engineering, Montreal, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
- Université de Montréal, Department of Medicine, Montreal, Canada
| | - Thomas Gervais
- Polytechnique Montréal, Department of Engineering Physics and Institute of Biomedical Engineering, Montreal, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Canada
| |
Collapse
|
22
|
St-Georges-Robillard A, Masse M, Cahuzac M, Strupler M, Patra B, Orimoto AM, Kendall-Dupont J, Péant B, Mes-Masson AM, Leblond F, Gervais T. Fluorescence hyperspectral imaging for live monitoring of multiple spheroids in microfluidic chips. Analyst 2019; 143:3829-3840. [PMID: 29999046 DOI: 10.1039/c8an00536b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tumor spheroids represent a realistic 3D in vitro cancer model because they provide a missing link between monolayer cell culture and live tissues. While microfluidic chips can easily form and assay thousands of spheroids simultaneously, few commercial instruments are available to analyze this massive amount of data. Available techniques to measure spheroid response to external stimuli, such as confocal imaging and flow cytometry, are either not appropriate for 3D cultures, or destructive. We designed a wide-field hyperspectral imaging system to analyze multiple spheroids trapped in a microfluidic chip in a single acquisition. The system and its fluorescence quantification algorithm were assessed using liquid phantoms mimicking spheroid optical properties. Spectral unmixing was tested on three overlapping spectral entities. Hyperspectral images of co-culture spheroids expressing two fluorophores were compared with confocal microscopy and spheroid growth was measured over time. The system can spectrally analyze multiple fluorescent markers simultaneously and allows multiple time-points assays, providing a fast and versatile solution for analyzing lab on a chip devices.
Collapse
Affiliation(s)
- Amélie St-Georges-Robillard
- Department of Engineering Physics, Polytechnique Montréal, C.P. 6079, Succ. Centre-ville, Montreal, Qc H3C 3A7, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Simeone K, Guay-Lord R, Lateef MA, Péant B, Kendall-Dupont J, Orimoto AM, Carmona E, Provencher D, Saad F, Gervais T, Mes-Masson AM. Paraffin-embedding lithography and micro-dissected tissue micro-arrays: tools for biological and pharmacological analysis of ex vivo solid tumors. LAB ON A CHIP 2019; 19:693-705. [PMID: 30671574 DOI: 10.1039/c8lc00982a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
There is an urgent need and strong clinical and pharmaceutical interest in developing assays that allow for the direct testing of therapeutic agents on primary tissues. Current technologies fail to provide the required sample longevity, throughput, and integration with standard clinically proven assays to make the approach viable. Here we report a microfluidic micro-histological platform that enables ex vivo culture of a large array of prostate and ovarian cancer micro-dissected tissue (MDT) followed by direct on-chip fixation and paraffination, a process we term paraffin-embedding lithography (PEL). The result is a high density MDT-Micro Array (MDTMA) compatible with standard clinical histopathology that can be used to analyse ex vivo tumor response or resistance to therapeutic agents. The cellular morphology and tissue architecture are preserved in MDTs throughout the 15 day culture period. We also demonstrate how this methodology can be used to study molecular pathways involved in cancer by performing in-depth characterization of biological and pharmacological mechanisms such as p65 nuclear translocation via TNF stimuli, and to predict the treatment outcome in the clinic via MDT response to taxane-based therapies.
Collapse
Affiliation(s)
- Kayla Simeone
- Centre de recherche du CHUM (CRCHUM)/Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gagné A, Têtu B, Orain M, Turcotte S, Plante M, Grégoire J, Renaud MC, Bairati I, Trueel D. HtrA1 expression and the prognosis of high-grade serous ovarian carcinoma: a cohort study using digital analysis. Diagn Pathol 2018; 13:57. [PMID: 30131069 PMCID: PMC6104006 DOI: 10.1186/s13000-018-0736-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/14/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The expression of high temperature requirement factor A1 (Htra1) has been reported to be decreased in ovarian carcinoma, but its prognostic effect remains undetermined. METHODS We evaluated the impact of HtrA1 downregulation in tumoral tissues on cancer progression and death in women with serous ovarian carcinoma. HtrA1 staining was performed on tissue microarrays (TMA) comprised of tumor samples from a cohort of 106 women who were diagnosed with primary high-grade serous ovarian carcinoma and receiving standard treatment at the Québec University Hospital between 1993 and 2006. HtrA1 expression was assessed visually (percentage of positive nuclei) and by digital image analysis (percentage of positive area). Cox regression multivariate models included standard prognostic factors and were used to estimate adjusted hazard ratios (aHR) for progression or death in the cohort. RESULTS By visual analysis, a low percentage of HtrA1-positive nuclei (< 10% vs ≥10%) tend to be associated with a lower risk of progression (aHR = 0.71; 95% Confidence interval (CI) = 0.46-1.09; P = 0.11) and mortality (aHR = 0.65; 95% CI = 0.41-1.04; P = 0.07). Low nuclear HtrA1 expression assessed by digital image analysis (< median % vs ≥ median %) showed a significant association with lower risk of progression (aHR = 0.62; 95% CI = 0.40-0.95; p = 0.03) and death (aHR = 0.60; 95% CI = 0.38-0.95; p = 0.03). CONCLUSION Altogether, our results demonstrate that nuclear downregulation of HtrA1 is associated with a better prognosis in women with high grade serous ovarian carcinoma.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/analysis
- Cell Nucleus/chemistry
- Cell Nucleus/pathology
- Cohort Studies
- Down-Regulation
- Female
- High-Temperature Requirement A Serine Peptidase 1/analysis
- Humans
- Image Interpretation, Computer-Assisted
- Immunohistochemistry
- Middle Aged
- Neoplasm Grading
- Neoplasms, Cystic, Mucinous, and Serous/chemistry
- Neoplasms, Cystic, Mucinous, and Serous/mortality
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Neoplasms, Cystic, Mucinous, and Serous/therapy
- Ovarian Neoplasms/chemistry
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/therapy
- Predictive Value of Tests
- Risk Assessment
- Risk Factors
- Time Factors
- Tissue Array Analysis
- Treatment Outcome
Collapse
Affiliation(s)
- Andréanne Gagné
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Bernard Têtu
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Anatomic Pathology and Cytology Department, Hôpital du St-Sacrement, Centre Hospitalier Universitaire (CHU) de Québec, Laval University, 1050 Chemin Ste-Foy, Québec, Québec G1S 4L8 Canada
- Department of Pathology, Hôpital du St-Sacrement, Centre Hospitalier Universitaire de Québec, 1050, Chemin Ste-Foy, Québec, Québec G1S 4L8 Canada
| | - Michèle Orain
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Anatomic Pathology and Cytology Department, Hôpital du St-Sacrement, Centre Hospitalier Universitaire (CHU) de Québec, Laval University, 1050 Chemin Ste-Foy, Québec, Québec G1S 4L8 Canada
| | - Stéphane Turcotte
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Marie Plante
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Gynecologic Oncology Division, Centre Hospitalier Universitaire (CHU) de Québec, L’Hôtel-Dieu-de-Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Jean Grégoire
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Gynecologic Oncology Division, Centre Hospitalier Universitaire (CHU) de Québec, L’Hôtel-Dieu-de-Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Marie-Claude Renaud
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Gynecologic Oncology Division, Centre Hospitalier Universitaire (CHU) de Québec, L’Hôtel-Dieu-de-Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Isabelle Bairati
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
| | - Dominique Trueel
- Laval University Cancer Research Center, Hôtel-Dieu-de-Québec, Centre Hospitalier Universitaire (CHU) de Québec, 11 Côte du Palais, Québec, Québec G1R 2J6 Canada
- Department of Pathology, Hôpital Saint-Luc, Centre Hospitalier Universitaire de Montréal, 058, rue Saint-Denis, Montréal, Québec H2X 3J4 Canada
- The Research Centre of the University of Montreal Teaching Hospital (CR-CHUM)/Montreal Cancer Institute, 900 Rue St-Denis, Montreal, Quebec H2X 0A9 Canada
- Department of Pathology and Cellular Biology, University of Montreal, 2900, boulevard Édouard-Montpetit, Montreal, Quebec H3T 1J4 Canada
| |
Collapse
|
25
|
Replication Protein A Availability during DNA Replication Stress Is a Major Determinant of Cisplatin Resistance in Ovarian Cancer Cells. Cancer Res 2018; 78:5561-5573. [DOI: 10.1158/0008-5472.can-18-0618] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/11/2018] [Accepted: 07/27/2018] [Indexed: 11/16/2022]
|
26
|
Fleury H, Carmona E, Morin VG, Meunier L, Masson JY, Tonin PN, Provencher D, Mes-Masson AM. Cumulative defects in DNA repair pathways drive the PARP inhibitor response in high-grade serous epithelial ovarian cancer cell lines. Oncotarget 2018; 8:40152-40168. [PMID: 27374179 PMCID: PMC5522225 DOI: 10.18632/oncotarget.10308] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/09/2016] [Indexed: 01/17/2023] Open
Abstract
PARP inhibitors (PARPi), such as Olaparib, have shown promising results in high-grade serous (HGS) epithelial ovarian cancer (EOC) treatment. PARPi sensitivity has been mainly associated with homologous recombination (HR) deficiency, but clinical trials have shown that predicting actual patient response is complex. Here, we investigated gene expression microarray, HR functionality and Olaparib sensitivity of 18 different HGS EOC cell lines and demonstrate that PARPi sensitivity is not only associated with HR defects. Gene target validation show that down regulation of genes in the nucleotide excision repair (NER) and mismatch repair (MMR) pathways (ERCC8 and MLH1, respectively) increases PARPi response. The highest sensitivity was observed when genes in both the HR and either NER or MMR pathways were concomitantly down regulated. Using clinical samples, patients with these concurrent down regulations could be identified. Based on these results, a novel model to predict PARPi sensitivity is herein proposed. This model implies that the extreme responders identified in clinical trials have deficiencies in HR and either NER or MMR.
Collapse
Affiliation(s)
- Hubert Fleury
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Euridice Carmona
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Vincent G Morin
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Liliane Meunier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU Research Center, Québec City, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University Cancer Research Center, Québec City, Canada
| | - Patricia N Tonin
- Cancer Research Program (CRP), The Research Institute of the McGill University Health Centre, Montreal, Canada.,Department of Human Genetics, McGill University, Montreal, Canada.,Department of Medicine, McGill University, Montreal, Canada
| | - Diane Provencher
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Division of Gynecologic Oncology, Université de Montréal, Montreal, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Department of Medicine, Université de Montréal, Montreal, Canada
| |
Collapse
|
27
|
Reconfigurable Microfluidic Magnetic Valve Arrays: Towards a Radiotherapy-Compatible Spheroid Culture Platform for the Combinatorial Screening of Cancer Therapies. SENSORS 2017; 17:s17102271. [PMID: 28976942 PMCID: PMC5677148 DOI: 10.3390/s17102271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 01/08/2023]
Abstract
We introduce here a microfluidic cell culture platform or spheroid culture chamber array (SCCA) that can synthesize, culture, and enable fluorescence imaging of 3D cell aggregates (typically spheroids) directly on-chip while specifying the flow of reagents in each chamber via the use of an array of passive magnetic valves. The SCCA valves demonstrated sufficient resistance to burst (above 100 mBar), including after receiving radiotherapy (RT) doses of up to 8 Gy combined with standard 37 °C incubation for up to 7 days, enabling the simultaneous synthesis of multiple spheroids from different cell lines on the same array. Our results suggest that SCCA would be an asset in drug discovery processes, seeking to identify combinatorial treatments.
Collapse
|
28
|
Teng PN, Bateman NW, Wang G, Litzi T, Blanton BE, Hood BL, Conrads KA, Ao W, Oliver KE, Darcy KM, McGuire WP, Paz K, Sidransky D, Hamilton CA, Maxwell GL, Conrads TP. Establishment and characterization of a platinum- and paclitaxel-resistant high grade serous ovarian carcinoma cell line. Hum Cell 2017; 30:226-236. [PMID: 28251557 DOI: 10.1007/s13577-017-0162-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/30/2017] [Indexed: 10/20/2022]
Abstract
High grade serous ovarian cancer (HGSOC) patients have a high recurrence rate after surgery and adjuvant chemotherapy due to inherent or acquired drug resistance. Cell lines derived from HGSOC tumors that are resistant to chemotherapeutic agents represent useful pre-clinical models for drug discovery. Here, we describe establishment of a human ovarian carcinoma cell line, which we term WHIRC01, from a patient-derived mouse xenograft established from a chemorefractory HGSOC patient who did not respond to carboplatin and paclitaxel therapy. This newly derived cell line is platinum- and paclitaxel-resistant with cisplatin, carboplatin, and paclitaxel half-maximal lethal doses of 15, 130, and 20 µM, respectively. Molecular characterization of this cell line was performed using targeted DNA exome sequencing, transcriptomics (RNA-seq), and mass spectrometry-based proteomic analyses. Results from exomic sequencing revealed mutations in TP53 consistent with HGSOC. Transcriptomic and proteomic analyses of WHIRC01 showed high level of alpha-enolase and vimentin, which are associated with cell migration and epithelial-mesenchymal transition. WHIRC01 represents a chemorefractory human HGSOC cell line model with a comprehensive molecular profile to aid future investigations of drug resistance mechanisms and screening of chemotherapeutic agents.
Collapse
Affiliation(s)
- Pang-Ning Teng
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Nicholas W Bateman
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA.,The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA.,Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Guisong Wang
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Tracy Litzi
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Brian E Blanton
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Brian L Hood
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Kelly A Conrads
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Wei Ao
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA
| | - Kate E Oliver
- Gynecologic Oncology Service, Department of Obstetrics and Gynecology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Kathleen M Darcy
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA.,The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - William P McGuire
- Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Keren Paz
- Champions Oncology, Inc., Baltimore, MD, USA
| | - David Sidransky
- Otolaryngology-Head and Neck Surgery and Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Chad A Hamilton
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA.,The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA.,Gynecologic Oncology Service, Department of Obstetrics and Gynecology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - G Larry Maxwell
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA.,The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA.,Department of Obstetrics and Gynecology, Inova Fairfax Hospital, Falls Church, VA, USA.,Inova Schar Cancer Institute, Inova Center for Personalized Health, Falls Church, VA, USA
| | - Thomas P Conrads
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA, USA. .,The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD, USA. .,Department of Obstetrics and Gynecology, Inova Fairfax Hospital, Falls Church, VA, USA. .,Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA. .,Inova Schar Cancer Institute, Inova Center for Personalized Health, Falls Church, VA, USA.
| |
Collapse
|
29
|
Borges A, Adega F, Chaves R. Establishment and characterization of a new feline mammary cancer cell line, FkMTp. Cytotechnology 2016; 68:1529-43. [PMID: 26883919 DOI: 10.1007/s10616-015-9912-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/14/2015] [Indexed: 12/28/2022] Open
Abstract
Studies on tumours in domestic animals are believed to greatly contribute to a better understanding of similar diseases in humans. Comparative studies have shown that feline mammary carcinomas share important features with human breast cancers, including a similar biological behaviour and histological appearance. In the present study we have established and characterized at different cellular levels one feline mammary cancer cell line, FkMTp, derived from a cat mammary carcinoma. The FkMTp cell line revealed to be a promising resource and tool to study tumour microevolution and all the mechanisms and processes involved in carcinogenesis from the tumour (primary culture) to the immortalized cell line. Several assays were conducted to assess the growth behaviour, differentiated morphology, anchorage independent growth in soft agar, wound-healing invasion and migration of the cell line across time (from the primary culture until the 160th passage). FkMTp revealed increased levels of anchorage independence, migration and invasion according to the course of time as well as different numbers of ploidy. These results demonstrate and validate the in vitro tumorigenicity of the FkMTp cell line. During the cell line establishment, it was cryopreserved approximately every six passages, including the tumour primary culture, allowing now the possibility to access almost any specific momento of the tumour progression.
Collapse
Affiliation(s)
- Ana Borges
- Laboratory of Cytogenomics and Animal Genomics (CAG), Department of Genetics and Biotechnology (DGB), Universityof Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Filomena Adega
- Laboratory of Cytogenomics and Animal Genomics (CAG), Department of Genetics and Biotechnology (DGB), Universityof Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Raquel Chaves
- Laboratory of Cytogenomics and Animal Genomics (CAG), Department of Genetics and Biotechnology (DGB), Universityof Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal.
| |
Collapse
|
30
|
The human organic cation transporter OCT1 mediates high affinity uptake of the anticancer drug daunorubicin. Sci Rep 2016; 6:20508. [PMID: 26861753 PMCID: PMC4748219 DOI: 10.1038/srep20508] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/05/2016] [Indexed: 01/11/2023] Open
Abstract
Anthracyclines such as daunorubicin are anticancer agents that are transported into cells, and exert cytotoxicity by blocking DNA metabolism. Although there is evidence for active uptake of anthracyclines into cells, the specific transporter involved in this process has not been identified. Using the high-grade serous ovarian cancer cell line TOV2223G, we show that OCT1 mediated the high affinity (Km ~ 5 μM) uptake of daunorubicin into the cells, and that micromolar amounts of choline completely abolished the drug entry. OCT1 downregulation by shRNA impaired daunorubicin uptake into the TOV2223G cells, and these cells were significantly more resistant to the drug in comparison to the control shRNA. Transfection of HEK293T cells, which accommodated the ectopic expression of OCT1, with a plasmid expressing OCT1-EYFP showed that the transporter was predominantly localized to the plasma membrane. These transfected cells exhibited an increase in the uptake of daunorubicin in comparison to control cells transfected with an empty EYFP vector. Furthermore, a variant of OCT1, OCT1-D474C-EYFP, failed to enhance daunorubicin uptake. This is the first report demonstrating that human OCT1 is involved in the high affinity transport of anthracyclines. We postulate that OCT1 defects may contribute to the resistance of cancer cells treated with anthracyclines.
Collapse
|
31
|
Fleury H, Communal L, Carmona E, Portelance L, Arcand SL, Rahimi K, Tonin PN, Provencher D, Mes-Masson AM. Novel high-grade serous epithelial ovarian cancer cell lines that reflect the molecular diversity of both the sporadic and hereditary disease. Genes Cancer 2015; 6:378-398. [PMID: 26622941 PMCID: PMC4633166 DOI: 10.18632/genesandcancer.76] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Few cell line models of epithelial ovarian cancer (EOC) have been developed for the high-grade serous (HGS) subtype, which is the most common and lethal form of gynaecological cancer. Here we describe the establishment of six new EOC cell lines spontaneously derived from HGS tumors (TOV2978G, TOV3041G and TOV3291G) or ascites (OV866(2), OV4453 and OV4485). Exome sequencing revealed somatic TP53 mutations in five of the cell lines. One cell line has a novel BRCA1 splice-site mutation, and another, a recurrent BRCA2 nonsense mutation, both of germline origin. The novel BRCA1 mutation induced abnormal splicing, mRNA instability, resulting in the absence of BRCA1 protein. None of the cell lines harbor mutations in KRAS or BRAF, which are characteristic of other EOC subtypes. SNP arrays showed that all of the cell lines exhibited structural chromosomal abnormalities, copy number alterations and regions of loss of heterozygosity, consistent with those described for HGS. Four cell lines were able to produce 3D-spheroids, two exhibited anchorage-independent growth, and three (including the BRCA1 and BRCA2 mutated cell lines) formed tumors in SCID mice. These novel HGS EOC cell lines and their detailed characterization provide new research tools for investigating the most common and lethal form of EOC.
Collapse
Affiliation(s)
- Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Laudine Communal
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Euridice Carmona
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Lise Portelance
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Suzanna L Arcand
- The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kurosh Rahimi
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Pathology, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Canada
| | - Patricia N Tonin
- The Research Institute of the McGill University Health Centre, Montreal, Canada.,Department of Human Genetics, McGill University, Montreal, Canada.,Department of Medicine, McGill University, Montreal, Canada
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Division of Gynecologic Oncology, Université de Montréal, Montreal, Canada.,Department of Medicine, Université de Montréal, Montreal, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Department of Medicine, Université de Montréal, Montreal, Canada
| |
Collapse
|
32
|
Alkema NG, Wisman GBA, van der Zee AGJ, van Vugt MATM, de Jong S. Studying platinum sensitivity and resistance in high-grade serous ovarian cancer: Different models for different questions. Drug Resist Updat 2015; 24:55-69. [PMID: 26830315 DOI: 10.1016/j.drup.2015.11.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/04/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022]
Abstract
High-grade serous ovarian cancer (HGSOC) has the highest mortality rate among all gynecological cancers. Patients are generally diagnosed in an advanced stage with the majority of cases displaying platinum resistant relapses. Recent genomic interrogation of large numbers of HGSOC patient samples indicated high complexity in terms of genetic aberrations, intra- and intertumor heterogeneity and underscored their lack of targetable oncogenic mutations. Sub-classifications of HGSOC based on expression profiles, termed 'differentiated', 'immunoreactive', 'mesenchymal' and 'proliferative', were shown to have prognostic value. In addition, in almost half of all HGSOC patients, a deficiency in homologous recombination (HR) was found that potentially can be targeted using PARP inhibitors. Developing precision medicine requires advanced experimental models. In the current review, we discuss experimental HGSOC models in which resistance to platinum therapy and the use of novel therapeutics can be carefully studied. Panels of better-defined primary cell lines need to be established to capture the full spectrum of HGSOC subtypes. Further refinement of cell lines is obtained with a 3-dimensional culture model mimicking the tumor microenvironment. Alternatively, ex vivo ovarian tumor tissue slices are used. For in vivo studies, larger panels of ovarian cancer patient-derived xenografts (PDXs) are being established, encompassing all expression subtypes. Ovarian cancer PDXs grossly retain tumor heterogeneity and clinical response to platinum therapy is preserved. PDXs are currently used in drug screens and as avatars for patient response. The role of the immune system in tumor responses can be assessed using humanized PDXs and immunocompetent genetically engineered mouse models. Dynamic tracking of genetic alterations in PDXs as well as patients during treatment and after relapse is feasible by sequencing circulating cell-free tumor DNA and analyzing circulating tumor cells. We discuss how various models and methods can be combined to delineate the molecular mechanisms underlying platinum resistance and to select HGSOC patients other than BRCA1/2-mutation carriers that could potentially benefit from the synthetic lethality of PARP inhibitors. This integrated approach is a first step to improve therapy outcomes in specific subgroups of HGSOC patients.
Collapse
Affiliation(s)
- Nicolette G Alkema
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ate G J van der Zee
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Steven de Jong
- Department of Medical Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
33
|
Kreuzinger C, Gamperl M, Wolf A, Heinze G, Geroldinger A, Lambrechts D, Boeckx B, Smeets D, Horvat R, Aust S, Hamilton G, Zeillinger R, Cacsire Castillo-Tong D. Molecular characterization of 7 new established cell lines from high grade serous ovarian cancer. Cancer Lett 2015; 362:218-28. [DOI: 10.1016/j.canlet.2015.03.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/23/2015] [Accepted: 03/31/2015] [Indexed: 12/27/2022]
|
34
|
Gambaro K, Quinn MCJ, Cáceres-Gorriti KY, Shapiro RS, Provencher D, Rahimi K, Mes-Masson AM, Tonin PN. Low levels of IGFBP7 expression in high-grade serous ovarian carcinoma is associated with patient outcome. BMC Cancer 2015; 15:135. [PMID: 25886299 PMCID: PMC4381406 DOI: 10.1186/s12885-015-1138-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 02/26/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Insulin-like growth factor binding protein 7 (IGFBP7) has been suggested to act as a tumour suppressor gene in various human cancers, yet its role in epithelial ovarian cancer (EOC) has not yet been investigated. We previously observed that IGFBP7 was one of several genes found significantly upregulated in an EOC cell line model rendered non-tumourigenic as consequence of genetic manipulation. The aim of the present study was to investigate the role of IGFBP7 in high-grade serous ovarian carcinomas (HGSC), the most common type of EOC. METHODS We analysed IGFBP7 gene expression in 11 normal ovarian surface epithelial cells (NOSE), 79 high-grade serous ovarian carcinomas (HGSC), and seven EOC cell lines using a custom gene expression array platform. IGFBP7 mRNA expression profiles were also extracted from publicly available databases. Protein expression was assessed by immunohistochemistry of 175 HGSC and 10 normal fallopian tube samples using tissue microarray and related to disease outcome. We used EOC cells to investigate possible mechanisms of gene inactivation and describe various in vitro growth effects of exposing EOC cell lines to human recombinant IGFBP7 protein and conditioned media. RESULTS All HGSCs exhibited IGFBP7 expression levels that were significantly (p = 0.001) lower than the mean of the expression value of NOSE samples and that of a whole ovary sample. IGFBP7 gene and protein expression were lower in tumourigenic EOC cell lines relative to a non-tumourigenic EOC cell line. None of the EOC cell lines harboured a somatic mutation in IGFBP7, although loss of heterozygosity (LOH) of the IGFBP7 locus and epigenetic methylation silencing of the IGFBP7 promoter was observed in two of the cell lines exhibiting loss of gene/protein expression. In vitro functional assays revealed an alteration of the EOC cell migration capacity. Protein expression analysis of HGSC samples revealed that the large majority of tumour cores (72.6%) showed low or absence of IGFBP7 staining and revealed a significant correlation between IGFBP7 protein expression and a prolonged overall survival (p = 0.044). CONCLUSION The low levels of IGFPB7 in HGSC relative to normal tissues, and association with survival are consistent with a purported role in tumour suppressor pathways.
Collapse
Affiliation(s)
- Karen Gambaro
- Department of Human Genetics, McGill University, Montreal, H3A 1B1, Canada. .,Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, H2X 0B9, Canada.
| | - Michael C J Quinn
- Department of Human Genetics, McGill University, Montreal, H3A 1B1, Canada. .,Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, H2X 0B9, Canada.
| | - Katia Y Cáceres-Gorriti
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, H2X 0B9, Canada.
| | - Rebecca S Shapiro
- Department of Human Genetics, McGill University, Montreal, H3A 1B1, Canada.
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, H2X 0B9, Canada. .,Department of Obstetric-Gynecology, Université de Montréal, Montreal, H2L 4M1, Canada.
| | - Kurosh Rahimi
- Department of Pathology, Université de Montréal, Montreal, H3C 3J7, Canada.
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, H2X 0B9, Canada. .,Department of Medicine, Université de Montréal, Montreal, H3C 3J7, Canada.
| | - Patricia N Tonin
- Department of Human Genetics, McGill University, Montreal, H3A 1B1, Canada. .,The Research Institute of the McGill University Health Centre, Montreal, H4A 3J1, Canada. .,Department of Medicine, McGill University, Montreal, H3G 1A4, Canada. .,Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Site Glen Pavillion Block E, Cancer Research Program E026217 (cubicle E), Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
35
|
Cunnea P, Stronach EA. Modeling platinum sensitive and resistant high-grade serous ovarian cancer: development and applications of experimental systems. Front Oncol 2014; 4:81. [PMID: 24860781 PMCID: PMC4029026 DOI: 10.3389/fonc.2014.00081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/02/2014] [Indexed: 12/29/2022] Open
Abstract
High-grade serous ovarian cancer remains the most common sub-type of ovarian cancer and, characterized by high degrees of genomic instability and heterogeneity, is typified by a transition from early response to acquired resistance to platinum-based chemotherapy. Conventional models for the study of ovarian cancer have been largely limited to a set of relatively poorly characterized immortalized cell lines and recent studies have called into question the validity of some of these as reliable models. Here, we review new approaches and models systems that take into account advances in our understanding of ovarian cancer biology and advances in the technology available for their generation and study. We discuss primary cell models, 2D, 3D, and organotypic models, and “paired” sample approaches that capture the evolution of chemotherapy failure within single cases. We also overview new methods for non-invasive collection of representative tumor material from blood samples. Adoption of such methods and models will improve the quality and clinical relevance of ovarian cancer research.
Collapse
Affiliation(s)
- Paula Cunnea
- Molecular Therapeutics Laboratory, Ovarian Cancer Action Research Centre, Institute of Reproductive and Developmental Biology, Department of Cancer and Surgery , Imperial College London, London , UK
| | - Euan A Stronach
- Molecular Therapeutics Laboratory, Ovarian Cancer Action Research Centre, Institute of Reproductive and Developmental Biology, Department of Cancer and Surgery , Imperial College London, London , UK
| |
Collapse
|
36
|
Garibay-Cerdenares OL, Hernández-Ramírez VI, Osorio-Trujillo JC, Hernández-Ortíz M, Gallardo-Rincón D, Cantú de León D, Encarnación-Guevara S, Villegas-Pineda JC, Talamás-Rohana P. Proteomic identification of fucosylated haptoglobin alpha isoforms in ascitic fluids and its localization in ovarian carcinoma tissues from Mexican patients. J Ovarian Res 2014; 7:27. [PMID: 24576319 PMCID: PMC3943579 DOI: 10.1186/1757-2215-7-27] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 02/19/2014] [Indexed: 12/18/2022] Open
Abstract
Background Ovarian cancer is the most lethal gynecologic disease due to delayed diagnosis, and ascites production is a characteristic of patients in advanced stages. The aim of this study was to perform the proteomic analysis of ascitic fluids of Mexican patients with ovarian carcinoma, in order to detect proteins with a differential expression pattern in the continuing search to identify biomarkers for this disease. Methods Samples were collected from 50 patients from the Instituto Nacional de Cancerología of México under informed consent and with approval of the bioethics and scientific committees. After elimination of abundant proteins (Albumin/IgGs) samples were processed for 2D electrophoresis and further protein identification by Mass Spectrometry (MALDI-TOF). Molecules of interest were followed by western blot and lectin binding assays, and their tissue location by histo-immunofluorescence and confocal analysis. Results and discussion An area with a differential expression pattern among samples was located in the 2D gels. Identified proteins were 6 alpha 1 isoforms and 1 alpha 2 isoform of Haptoglobin, and 2 isoforms of Transthyretin. While Transthyretin isoforms were constitutively expressed in all samples, clear differences in the expression pattern of Haptoglobin alpha isoforms were found. Moreover, increased levels of fucosylation of Haptoglobin alpha isoforms analyzed in 40 samples by Aleuria aurantia lectin binding by 1D overlay assay showed a positive correlation with advanced stages of the disease. Tissue detection of Haptoglobin and its fucosylated form, by histo-immunofluorescence in biopsies of ovarian cancer, also showed a correlation with ovarian cancer progression. Moreover, results show that fucosylated Haptoglobin is produced by tumor cells. Conclusions Increased numbers of highly fucosylated Haptoglobin alpha isoforms in ascitic fluids and the presence of fucosylated Haptoglobin in tumor tissues of ovarian cancer Mexican patients associated with advanced stages of the disease, reinforce the potential of fucosylated Haptoglobin alpha isoforms to be characterized as biomarkers for disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Patricia Talamás-Rohana
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av, Instituto Politécnico Nacional 2508, Col, San Pedro Zacatenco, Delegación Gustavo A, Madero, México, D,F, 07360, México.
| |
Collapse
|
37
|
Granulocyte colony-stimulating factor receptor signalling via Janus kinase 2/signal transducer and activator of transcription 3 in ovarian cancer. Br J Cancer 2013; 110:133-45. [PMID: 24220695 PMCID: PMC3887286 DOI: 10.1038/bjc.2013.673] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/25/2013] [Accepted: 10/04/2013] [Indexed: 12/24/2022] Open
Abstract
Background: Ovarian cancer remains a major cause of cancer mortality in women, with only limited understanding of disease aetiology at the molecular level. Granulocyte colony-stimulating factor (G-CSF) is a key regulator of both normal and emergency haematopoiesis, and is used clinically to aid haematopoietic recovery following ablative therapies for a variety of solid tumours including ovarian cancer. Methods: The expression of G-CSF and its receptor, G-CSFR, was examined in primary ovarian cancer samples and a panel of ovarian cancer cell lines, and the effects of G-CSF treatment on proliferation, migration and survival were determined. Results: G-CSFR was predominantly expressed in high-grade serous ovarian epithelial tumour samples and a subset of ovarian cancer cell lines. Stimulation of G-CSFR-expressing ovarian epithelial cancer cells with G-CSF led to increased migration and survival, including against chemotherapy-induced apoptosis. The effects of G-CSF were mediated by signalling via the downstream JAK2/STAT3 pathway. Conclusion: This study suggests that G-CSF has the potential to impact on ovarian cancer pathogenesis, and that G-CSFR expression status should be considered in determining appropriate therapy.
Collapse
|
38
|
Quinn MCJ, Wojnarowicz PM, Pickett A, Provencher DM, Mes-Masson AM, Davis EC, Tonin PN. FKBP10/FKBP65 expression in high-grade ovarian serous carcinoma and its association with patient outcome. Int J Oncol 2013; 42:912-20. [PMID: 23354471 DOI: 10.3892/ijo.2013.1797] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 12/05/2012] [Indexed: 11/06/2022] Open
Abstract
The frequent loss of chromosome 17 in epithelial ovarian carcinomas (EOC), particularly high-grade serous carcinomas (HGSC), has been attributed to the disruption of TP53 (at 17p13.1) and other chromosome 17 genes suspected to play a role in tumour suppressor pathways. In a transcriptome analysis of HGSC, we showed underexpression of a number of chromosome 17 genes, which included FKBP10 (at 17q21.1) and collagen I α 1 (COL1A1; at 17q21.33). FKBP10 codes for the immunophilin FKBP65 and is suspected to act as a chaperone for COL1A1. We have investigated FKBP10 (gene) and FKBP65 (protein) expression in HGSC samples and EOC cell lines that differ in their tumourigenic potential. COL1A1 expression was also investigated given the purported function of FKBP65. RT-PCR analysis verified underexpression of FKBP10 and COL1A1 in HGSCs (n=14) and six tumourigenic EOC cell lines, relative to normal ovarian surface epithelial cells and a non-tumourigenic EOC cell line. Immunohistochemistry analyses of 196 HGSC samples using tissue microarrays revealed variable staining intensities in the epithelial tumour component where only 7.8% and 1.0% of samples stained intensely for FKBP65 and COL1A1, respectively. Variable staining intensities were also observed for the stromal component where 23.6% and 24.1% stained intensely for FKBP65 and COL1A1, respectively. There was no significant correlation of staining intensity of either protein with disease stage. Staining of FKBP65 was clearly visible in normal epithelial cells of the ovarian surface and fallopian tube. There was a significant correlation between absence of FKBP65 staining in the epithelial cell component of the tumour and prolonged overall survival (p<0.001). Our results suggest that underexpression of FKBP65 protein is characteristic of HGSCs and that this expression profile may be linked to molecular pathways associated with an unfavourable outcome in cancer patients.
Collapse
Affiliation(s)
- Michael C J Quinn
- Research Centre of the University of Montreal Hospital Centre/Montreal Cancer Institute, Montreal, QC, Canada
| | | | | | | | | | | | | |
Collapse
|
39
|
Establishment of primary cultures from ovarian tumor tissue and ascites fluid. Methods Mol Biol 2013; 1049:323-36. [PMID: 23913227 DOI: 10.1007/978-1-62703-547-7_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
We have refined the technique for isolating and propagating cultures of primary epithelial ovarian cancer (EOC) cells derived from solid tumors and ascites. Both protocols involve a simple yet rapid method for the growth and propagation of EOC tumor and ascites cells in a basal culture medium without the addition of growth factors. Isolation of tumor EOC cells involves the mechanical disruption of the tumor tissue with the help of a cell scraper, while ascites-derived EOC cells are mixed with growth medium and placed directly into culture with very little manipulation. We further describe a partial trypsinization method to eliminate fibroblast contamination from primary EOC cells derived from solid tumors. These methods allow for the direct application of many molecular, cellular, and functional analyses within a few weeks of initial isolation, with the added potential of retrospective analyses of archived cells and tissues. Thus, we have included steps for long-term cryopreservation of early-passage EOC cells. Initial isolation of EOC cells can be completed within 1 h, and primary cells are further expanded in culture for several weeks.
Collapse
|
40
|
Das T, Meunier L, Barbe L, Provencher D, Guenat O, Gervais T, Mes-Masson AM. Empirical chemosensitivity testing in a spheroid model of ovarian cancer using a microfluidics-based multiplex platform. BIOMICROFLUIDICS 2013; 7:11805. [PMID: 24403987 PMCID: PMC3555942 DOI: 10.1063/1.4774309] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/18/2012] [Indexed: 05/11/2023]
Abstract
The use of biomarkers to infer drug response in patients is being actively pursued, yet significant challenges with this approach, including the complicated interconnection of pathways, have limited its application. Direct empirical testing of tumor sensitivity would arguably provide a more reliable predictive value, although it has garnered little attention largely due to the technical difficulties associated with this approach. We hypothesize that the application of recently developed microtechnologies, coupled to more complex 3-dimensional cell cultures, could provide a model to address some of these issues. As a proof of concept, we developed a microfluidic device where spheroids of the serous epithelial ovarian cancer cell line TOV112D are entrapped and assayed for their chemoresponse to carboplatin and paclitaxel, two therapeutic agents routinely used for the treatment of ovarian cancer. In order to index the chemoresponse, we analyzed the spatiotemporal evolution of the mortality fraction, as judged by vital dyes and confocal microscopy, within spheroids subjected to different drug concentrations and treatment durations inside the microfluidic device. To reflect microenvironment effects, we tested the effect of exogenous extracellular matrix and serum supplementation during spheroid formation on their chemotherapeutic response. Spheroids displayed augmented chemoresistance in comparison to monolayer culturing. This resistance was further increased by the simultaneous presence of both extracellular matrix and high serum concentration during spheroid formation. Following exposure to chemotherapeutics, cell death profiles were not uniform throughout the spheroid. The highest cell death fraction was found at the center of the spheroid and the lowest at the periphery. Collectively, the results demonstrate the validity of the approach, and provide the basis for further investigation of chemotherapeutic responses in ovarian cancer using microfluidics technology. In the future, such microdevices could provide the framework to assay drug sensitivity in a timeframe suitable for clinical decision making.
Collapse
Affiliation(s)
- Tamal Das
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montréal, Québec H2L 4M1, Canada
| | - Liliane Meunier
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montréal, Québec H2L 4M1, Canada
| | - Laurent Barbe
- Centre Suisse d' Electronique et de Microtechnique, CH-7302 Landquart, Switzerland
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montréal, Québec H2L 4M1, Canada ; Division of Gynecologic Oncology, Université de Montréal, Montréal, Québec H2L 4M1, Canada
| | - Olivier Guenat
- ARTORG Center, University of Bern, CH-3010 Bern, Switzerland
| | - Thomas Gervais
- Department of Engineering Physics, École Polytechnique de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montréal, Québec H2L 4M1, Canada ; Département de médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
41
|
Wojnarowicz PM, Oros KK, Quinn MCJ, Arcand SL, Gambaro K, Madore J, Birch AH, de Ladurantaye M, Rahimi K, Provencher DM, Mes-Masson AM, Greenwood CMT, Tonin PN. The genomic landscape of TP53 and p53 annotated high grade ovarian serous carcinomas from a defined founder population associated with patient outcome. PLoS One 2012; 7:e45484. [PMID: 23029043 PMCID: PMC3447752 DOI: 10.1371/journal.pone.0045484] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/16/2012] [Indexed: 11/20/2022] Open
Abstract
High-grade ovarian serous carcinomas (HGSC) are characterized by TP53 mutations and non-random patterns of chromosomal anomalies, where the nature of the TP53 mutation may correlate with clinical outcome. However, the frequency of common somatic genomic events occurring in HGSCs from demographically defined populations has not been explored. Whole genome SNP array, and TP53 mutation, gene and protein expression analyses were assessed in 87 confirmed HGSC samples with clinical correlates from French Canadians, a population exhibiting strong founder effects, and results were compared with independent reports describing similar analyses from unselected populations. TP53 mutations were identified in 91% of HGSCs. Anomalies observed in more than 50% of TP53 mutation-positive HGSCs involved gains of 3q, 8q and 20q, and losses of 4q, 5q, 6q, 8p, 13q, 16q, 17p, 17q, 22q and Xp. Nearly 400 regions of non-overlapping amplification or deletion were identified, where 178 amplifications and 98 deletions involved known genes. The subgroup expressing mutant p53 protein exhibited significantly prolonged overall and disease-free survival as compared with the p53 protein null subgroup. Interestingly, a comparative analysis of genomic landscapes revealed a significant enrichment of gains involving 1q, 8q, and 12p intervals in the subgroup expressing mutant p53 protein as compared with the p53 protein null subgroup. Although the findings show that the frequency of TP53 mutations and the genomic landscapes observed in French Canadian samples were similar to those reported for samples from unselected populations, there were differences in the magnitude of global gains/losses of specific chromosomal arms and in the spectrum of amplifications and deletions involving focal regions in individual samples. The findings from our comparative genomic analyses also support the notion that there may be biological differences between HGSCs that could be related to the nature of the TP53 mutation.
Collapse
Affiliation(s)
| | - Kathleen Klein Oros
- Division of Clinical Epidemiology and Segal Cancer Centre, Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec, Canada
| | - Michael C. J. Quinn
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Suzanna L. Arcand
- The Research Institute of the McGill University Health Centre (MUHC), Montreal, Quebec, Canada
| | - Karen Gambaro
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Jason Madore
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Ashley H. Birch
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Manon de Ladurantaye
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montreal, Quebec, Canada
| | - Kurosh Rahimi
- Department of Pathology, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Diane M. Provencher
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montreal, Quebec, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montreal, Quebec, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Institut du Cancer de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Celia M. T. Greenwood
- Division of Clinical Epidemiology and Segal Cancer Centre, Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Oncology, McGill University, Montreal, Quebec, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Patricia N. Tonin
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- The Research Institute of the McGill University Health Centre (MUHC), Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Wojnarowicz P, Gambaro K, de Ladurantaye M, Quinn MCJ, Provencher D, Mes-Masson AM, Tonin PN. Overexpressing the CCL2 chemokine in an epithelial ovarian cancer cell line results in latency of in vivo tumourigenicity. Oncogenesis 2012; 1:e27. [PMID: 23552840 PMCID: PMC3503293 DOI: 10.1038/oncsis.2012.25] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The frequent loss of heterozygosity of chromosome (Chr) 17 in epithelial ovarian cancer (EOC), particularly high-grade ovarian serous carcinomas (HGOSCs), has been attributed to the disruption of known tumour suppressor genes, such as TP53 (17p13), as well as other genes on this chromosome that alone or in combination have a role in EOC. In a transcriptome analysis of Chr17 genes, we observed significant underexpression of the chemokine CCL2 (17q12) in a small set of HGOSC samples relative to normal ovarian surface epithelial cells and a significant upregulation of CCL2 in the TP53-mutated OV-90 EOC cell line rendered non-tumourigenic as a consequence of genetic manipulation. Here, we report that overexpressing CCL2 in OV-90 resulted in latency of tumour formation at intraperitoneal (i.p.) but not subcutaneous sites in a mouse xenograft model. Overexpressing CCL2 affected cell morphology and exerted modest, but not significant effects on cell viability, colony formation and cell migration. We report significant underexpression of CCL2 by transcriptome analysis (P=0.015) and by immunohistochemistry in 77% of HGOSC samples (n=65). Absent or a very low level of protein expression by immunohistochemistry was also observed in 71% of additional HGOSC samples (n=122). However, CCL2 protein expression did not significantly correlate with overall or disease-free survival. The epithelial cells of normal fallopian tubes, a purported origin of HGOSC, exhibited expression of CCL2 protein by immunohistochemistry. Our results affirm that CCL2 underexpression is a significant feature of HGOSC samples, and that CCL2 overexpression in an EOC cell line model affects tumourigenic potential in the i.p. setting.
Collapse
Affiliation(s)
- P Wojnarowicz
- Department of Human Genetics, McGill University, Montreal, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Létourneau IJ, Quinn MCJ, Wang LL, Portelance L, Caceres KY, Cyr L, Delvoye N, Meunier L, de Ladurantaye M, Shen Z, Arcand SL, Tonin PN, Provencher DM, Mes-Masson AM. Derivation and characterization of matched cell lines from primary and recurrent serous ovarian cancer. BMC Cancer 2012; 12:379. [PMID: 22931248 PMCID: PMC3532154 DOI: 10.1186/1471-2407-12-379] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 08/20/2012] [Indexed: 01/02/2023] Open
Abstract
Background Cell line models have proven to be effective tools to investigate a variety of ovarian cancer features. Due to the limited number of cell lines, particularly of the serous subtype, the heterogeneity of the disease, and the lack of cell lines that model disease progression, there is a need to further develop cell line resources available for research. This study describes nine cell lines derived from three ovarian cancer cases that were established at initial diagnosis and at subsequent relapse after chemotherapy. Methods The cell lines from three women diagnosed with high-grade serous ovarian cancer (1369, 2295 and 3133) were derived from solid tumor (TOV) and ascites (OV), at specific time points at diagnosis and relapse (R). Primary treatment was a combination of paclitaxel/carboplatin (1369, 3133), or cisplatin/topotecan (2295). Second line treatment included doxorubicin, gemcitabine and topotecan. In addition to molecular characterization (p53, HER2), the cell lines were characterized based on cell growth characteristics including spheroid growth, migration potential, and anchorage independence. The in vivo tumorigenicity potential of the cell lines was measured. Response to paclitaxel and carboplatin was assessed using a clonogenic assay. Results All cell lines had either a nonsense or missense TP53 mutations. The ability to form compact spheroids or aggregates was observed in six of nine cell lines. Limited ability for migration and anchorage independence was observed. The OV3133(R) cell line, formed tumors at subcutaneous sites in SCID mice. Based on IC50 values and dose response curves, there was clear evidence of acquired resistance to carboplatin for TOV2295(R) and OV2295(R2) cell lines. Conclusion The study identified nine new high-grade serous ovarian cancer cell lines, derived before and after chemotherapy that provides a unique resource for investigating the evolution of this common histopathological subtype of ovarian cancer.
Collapse
Affiliation(s)
- Isabelle J Létourneau
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM)/Institut du cancer de Montréal, Montréal, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Birch AH, Arcand SL, Oros KK, Rahimi K, Watters AK, Provencher D, Greenwood CM, Mes-Masson AM, Tonin PN. Chromosome 3 anomalies investigated by genome wide SNP analysis of benign, low malignant potential and low grade ovarian serous tumours. PLoS One 2011; 6:e28250. [PMID: 22163003 PMCID: PMC3232202 DOI: 10.1371/journal.pone.0028250] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/04/2011] [Indexed: 02/01/2023] Open
Abstract
Ovarian carcinomas exhibit extensive heterogeneity, and their etiology remains unknown. Histological and genetic evidence has led to the proposal that low grade ovarian serous carcinomas (LGOSC) have a different etiology than high grade carcinomas (HGOSC), arising from serous tumours of low malignant potential (LMP). Common regions of chromosome (chr) 3 loss have been observed in all types of serous ovarian tumours, including benign, suggesting that these regions contain genes important in the development of all ovarian serous carcinomas. A high-density genome-wide genotyping bead array technology, which assayed >600,000 markers, was applied to a panel of serous benign and LMP tumours and a small set of LGOSC, to characterize somatic events associated with the most indolent forms of ovarian disease. The genomic patterns inferred were related to TP53, KRAS and BRAF mutations. An increasing frequency of genomic anomalies was observed with pathology of disease: 3/22 (13.6%) benign cases, 40/53 (75.5%) LMP cases and 10/11 (90.9%) LGOSC cases. Low frequencies of chr3 anomalies occurred in all tumour types. Runs of homozygosity were most commonly observed on chr3, with the 3p12-p11 candidate tumour suppressor region the most frequently homozygous region in the genome. An LMP harboured a homozygous deletion on chr6 which created a GOPC-ROS1 fusion gene, previously reported as oncogenic in other cancer types. Somatic TP53, KRAS and BRAF mutations were not observed in benign tumours. KRAS-mutation positive LMP cases displayed significantly more chromosomal aberrations than BRAF-mutation positive or KRAS and BRAF mutation negative cases. Gain of 12p, which harbours the KRAS gene, was particularly evident. A pathology review reclassified all TP53-mutation positive LGOSC cases, some of which acquired a HGOSC status. Taken together, our results support the view that LGOSC could arise from serous benign and LMP tumours, but does not exclude the possibility that HGOSC may derive from LMP tumours.
Collapse
Affiliation(s)
- Ashley H. Birch
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Suzanna L. Arcand
- The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kathleen K. Oros
- Division of Clinical Epidemiology and Segal Cancer Centre, Lady Davis Research Institute, Jewish General Hospital, Montreal, Canada
| | - Kurosh Rahimi
- Department of Pathology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Canada
| | - A. Kevin Watters
- Department of Pathology, McGill University and McGill University Health Centre (MUHC), Montréal, Canada
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Institut du cancer de Montréal, Montreal, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montreal, Canada
| | - Celia M. Greenwood
- Division of Clinical Epidemiology and Segal Cancer Centre, Lady Davis Research Institute, Jewish General Hospital, Montreal, Canada
- Department of Oncology, McGill University, Montreal, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Institut du cancer de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Patricia N. Tonin
- Department of Human Genetics, McGill University, Montreal, Canada
- The Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Medicine, McGill University, Montreal, Canada
- * E-mail:
| |
Collapse
|
45
|
Yoffou PH, Edjekouane L, Meunier L, Tremblay A, Provencher DM, Mes-Masson AM, Carmona E. Subtype specific elevated expression of hyaluronidase-1 (HYAL-1) in epithelial ovarian cancer. PLoS One 2011; 6:e20705. [PMID: 21695196 PMCID: PMC3112150 DOI: 10.1371/journal.pone.0020705] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 05/08/2011] [Indexed: 12/31/2022] Open
Abstract
Background Epithelial ovarian cancer (EOC) is morphologically heterogeneous being classified as serous, endometrioid, clear cell, or mucinous. Molecular genetic analysis has suggested a role for tumor suppressor genes located at chromosome 3p in serous EOC pathogenesis. Our objective was to evaluate the expression of HYAL1, located at chromosome 3p21.3, in these EOC subtypes, and to investigate its correlation with the expression of steroid hormone receptors. Methodology/Principal Findings We determined the mRNA expression of HYAL1, estrogen receptor (ER)-α, ERβ and progesterone receptor (PR) in EOC tumor samples and cell lines using quantitative RT-PCR. We also examined the expression of these genes in a publicly available microarray dataset. HYAL-1 enzyme activity was measured in EOC cell lines and in plasma samples from patients. We found that HYAL1 mRNA expression was elevated in clear cell and mucinous EOC tissue samples, but not in serous and endometrioid samples, normal ovaries or benign tumors. Similar results were obtained by two different techniques and with tissue sample cohorts from two independent institutions. Concordantly, HYAL1 mRNA levels and enzymatic activity were elevated only in EOC cell lines derived from clear cell and mucinous subtypes. We also showed that HYAL1 mRNA was inversely correlated to that of ERα specifically in clear cell and mucinous EOCs. Additionally, ectopic expression of ERα in a clear cell EOC cell line (ER- and PR-negative) induced 50% reduction of HYAL1 mRNA expression, supporting a role of ERα in HYAL1 gene regulation. Significantly, HYAL-1 activity was also high in the plasma of patients with these EOC subtypes. Conclusions/Significance This is the first report showing high HYAL-1 levels in EOC and demonstrating HYAL1 gene repression by ERα. Our results identify Hyaluronidase-1 as a potential target/biomarker for clear cell and mucinous EOCs and especially in tumors with low ERα levels.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/blood
- Adenocarcinoma, Clear Cell/enzymology
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Mucinous/blood
- Adenocarcinoma, Mucinous/enzymology
- Adenocarcinoma, Mucinous/pathology
- Biomarkers, Tumor/blood
- Carcinoma, Ovarian Epithelial
- Cell Line, Tumor
- Culture Media, Conditioned
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hyaluronoglucosaminidase/genetics
- Hyaluronoglucosaminidase/metabolism
- Neoplasms, Glandular and Epithelial/blood
- Neoplasms, Glandular and Epithelial/classification
- Neoplasms, Glandular and Epithelial/enzymology
- Neoplasms, Glandular and Epithelial/genetics
- Ovarian Neoplasms/blood
- Ovarian Neoplasms/classification
- Ovarian Neoplasms/enzymology
- Ovarian Neoplasms/genetics
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
Collapse
Affiliation(s)
| | - Lydia Edjekouane
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada
| | - Liliane Meunier
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - André Tremblay
- Department of Obstetrics and Gynecology, University of Montreal, Montreal, Quebec, Canada
- Department of Biochemistry, University of Montreal, Montreal, Quebec, Canada
- Research Center, Centre hospitalier universitaire Ste-Justine, Montreal, Quebec, Canada
| | - Diane Michèle Provencher
- Department of Obstetrics and Gynecology, University of Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Anne-Marie Mes-Masson
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Department of Obstetrics and Gynecology, University of Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Euridice Carmona
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
46
|
Kurbacher CM, Korn C, Dexel S, Schween U, Kurbacher JA, Reichelt R, Arenz PN. Isolation and culture of ovarian cancer cells and cell lines. Methods Mol Biol 2011; 731:161-180. [PMID: 21516407 DOI: 10.1007/978-1-61779-080-5_15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Ovarian carcinomas show considerable heterogeneity of origin, both in terms of site and tissue. The most important and also most frequent of these tumors arise from the coelomic epithelium and are therefore characterized as epithelial ovarian carcinomas (EOC). EOC is often large and advanced at the time of presentation, so that cells are readily obtainable from surgical specimens or effusions. While the primary tumor may be chemosensitive, they often develop resistance and may do so rapidly. Due to the easy access to tumor cells and its biological behavior, EOC is considered to be an ideal model to investigate principal mechanisms of both antineoplastic drug sensitivity and resistance. Although studies on primary EOC cells are now preferred for many of these investigations, EOC cell line studies remain important too. This chapter gives an overview over major techniques required to establish and maintain primary EOC cell cultures and to initiate and cultivate permanently growing EOC cell lines.
Collapse
|
47
|
Barrès V, Ouellet V, Lafontaine J, Tonin PN, Provencher DM, Mes-Masson AM. An essential role for Ran GTPase in epithelial ovarian cancer cell survival. Mol Cancer 2010; 9:272. [PMID: 20942967 PMCID: PMC2964620 DOI: 10.1186/1476-4598-9-272] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 10/13/2010] [Indexed: 12/03/2022] Open
Abstract
Background We previously identified that Ran protein, a member of the Ras GTPase family, is highly expressed in high grade and high stage serous epithelial ovarian cancers, and that its overexpression is associated with a poor prognosis. Ran is known to contribute to both nucleocytoplasmic transport and cell cycle progression, but its role in ovarian cancer is not well defined. Results Using a lentivirus-based tetracycline-inducible shRNA approach, we show that downregulation of Ran expression in aggressive ovarian cancer cell lines affects cellular proliferation by inducing a caspase-3 associated apoptosis. Using a xenograft tumor assay, we demonstrate that depletion of Ran results in decreased tumorigenesis, and eventual tumor formation is associated with tumor cells that express Ran protein. Conclusion Our results suggest a role for Ran in ovarian cancer cell survival and tumorigenicity and suggest that this critical GTPase may be suitable as a therapeutic target.
Collapse
Affiliation(s)
- Véronique Barrès
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal/Institut du cancer de Montréal,1560 Sherbrooke Est, Montreal H2L 4M1, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Effect of ovarian cancer ascites on cell migration and gene expression in an epithelial ovarian cancer in vitro model. Transl Oncol 2010; 3:230-8. [PMID: 20689764 DOI: 10.1593/tlo.10103] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 01/29/2010] [Accepted: 02/08/2010] [Indexed: 01/02/2023] Open
Abstract
A third of patients with epithelial ovarian cancer (EOC) present ascites. The cellular fraction of ascites often consists of EOC cells, lymphocytes, and mesothelial cells, whereas the acellular fraction contains cytokines and angiogenic factors. Clinically, the presence of ascites correlates with intraperitoneal and retroperitoneal tumor spread. We have used OV-90, a tumorigenic EOC cell line derived from the malignant ascites of a chemonaive ovarian cancer patient, as a model to assess the effect of ascites on migration potential using an in vitro wound-healing assay. A recent report of an invasion assay described the effect of ascites on the invasion potential of the OV-90 cell line. Ascites sampled from 31 ovarian cancer patients were tested and compared with either 5% fetal bovine serum or no serum for their nonstimulatory or stimulatory effect on the migration potential of the OV-90 cell line. A supervised analysis of data generated by the Affymetrix HG-U133A GeneChip identified differentially expressed genes from OV-90 cells exposed to ascites that had either a nonstimulatory or a stimulatory effect on migration. Ten genes (IRS2, CTSD, NRAS, MLXIP, HMGCR, LAMP1, ETS2, NID1, SMARCD1, and CD44) were upregulated in OV-90 cells exposed to ascites, allowing a nonstimulatory effect on cell migration. These findings were validated by quantitative polymerase chain reaction. In addition, the gene expression of IRS2 and MLXIP each correlated with prognosis when their expression was assessed in an independent set of primary cultures established from ovarian ascites. This study revealed novel candidates that may play a role in ovarian cancer cell migration.
Collapse
|
49
|
Abstract
Resistance to chemotherapy in ovarian cancer is poorly understood. Evolutionary models of cancer predict that, following treatment, resistance emerges either because of outgrowth of an intrinsically resistant sub-clone or evolves in residual disease under the selective pressure of treatment. To investigate genetic evolution in high-grade serous (HGS) ovarian cancers, we first analysed cell line series derived from three cases of HGS carcinoma before and after platinum resistance had developed (PEO1, PEO4 and PEO6; PEA1 and PEA2; and PEO14 and PEO23). Analysis with 24-colour fluorescence in situ hybridisation and single nucleotide polymorphism (SNP) array comparative genomic hybridisation (CGH) showed mutually exclusive endoreduplication and loss of heterozygosity events in clones present at different time points in the same individual. This implies that platinum-sensitive and -resistant disease was not linearly related, but shared a common ancestor at an early stage of tumour development. Array CGH analysis of six paired pre- and post-neoadjuvant treatment HGS samples from the CTCR-OV01 clinical study did not show extensive copy number differences, suggesting that one clone was strongly dominant at presentation. These data show that cisplatin resistance in HGS carcinoma develops from pre-existing minor clones but that enrichment for these clones is not apparent during short-term chemotherapy treatment.
Collapse
|
50
|
Protease inhibitor SERPINA1 expression in epithelial ovarian cancer. Clin Exp Metastasis 2010; 27:55-69. [PMID: 20049513 DOI: 10.1007/s10585-009-9303-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 12/08/2009] [Indexed: 12/23/2022]
Abstract
Epithelial ovarian cancer is the most lethal gynecologic cancer with a 5 years survival rate of 30-40% in patients diagnosed with high-grade invasive disease (TOV). This is in stark contrast to the 95% 5 years survival rate in ovarian cancer patients diagnosed with low malignant potential (LMP) disease. The progression from localized tumor to invasive metastasis involves matrix proteolysis. Protease inhibitors are thought to play a key role by limiting this process. Using the Affymetrix HG-U133A GeneChip array, we have studied all serine protease inhibitors and found several serpin family members that are differentially expressed between LMP and TOV serous tumors. SERPINA1 was selected for further study due to its high expression in the majority of LMP tumors and its low expression in TOV tumors; observations that were also validated by quantitative-PCR (Q-PCR). To study the effects of its over expression on different tumorigenic parameters, SERPINA1 was cloned in the pcDNA3.1+ plasmid which was subsequently used to derive stable clones from two invasive ovarian cancer cell lines, TOV-112D and TOV-1946. We found no effect of SERPINA1 over expression on tumor growth in SCID mice although cell migration and invasion were affected in in vitro assays. There was also no association between patient survival and SERPINA1 immunostaining, however, SERPINA1 localization was different in LMP (nuclear) and TOV (cytoplasmic) tumors. SERPINA1 remains an interesting candidate since protein homeostasis, regulated by proteases and their inhibitors, should be studied holistically in order to assess their full impact in tumor progression.
Collapse
|