101
|
Ihlow J, Monjé N, Hoffmann I, Bischoff P, Sinn BV, Schmitt WD, Kunze CA, Darb-Esfahani S, Kulbe H, Braicu EI, Sehouli J, Denkert C, Horst D, Taube ET. Low Expression of RGS2 Promotes Poor Prognosis in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14194620. [PMID: 36230542 PMCID: PMC9561967 DOI: 10.3390/cancers14194620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/03/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Recent advances in molecular medicine have indicated G-protein coupled receptors (GPCRs) as possible therapeutic targets in ovarian cancer. The cellular effects of GPCRs are determined by regulator of G protein signaling (RGS) proteins. Especially RGS2 has currently moved into focus of cancer therapy. Therefore, we retrospectively analyzed RGS2 and its association with the prognosis of high-grade serous ovarian cancer (HGSOC). Here, we provide in situ and in silico analyses regarding the expression patterns and prognostic value of RGS2. In silico we found that RGS2 is barely detectable in tumor cells on the mRNA level in bulk and single-cell data. Applying immunohistochemistry in 519 HGSOC patients, we detected moderate to strong protein expression of RGS2 in situ in approximately half of the cohort, suggesting regulation by post translational modification. Furthermore, low protein expression of RGS2 was associated with an inferior overall- and progression-free survival. These results warrant further research of its role and related new therapeutic implications in HGSOC. Abstract RGS2 regulates G-protein signaling by accelerating hydrolysis of GTP and has been identified as a potentially druggable target in carcinomas. Since the prognosis of patients with high-grade serous ovarian carcinoma (HGSOC) remains utterly poor, new therapeutic options are urgently needed. Previous in vitro studies have linked RGS2 suppression to chemoresistance in HGSOC, but in situ data are still missing. In this study, we characterized the expression of RGS2 and its relation to prognosis in HGSOC on the protein level by immunohistochemistry in 519 patients treated at Charité, on the mRNA level in 299 cases from TCGA and on the single-cell level in 19 cases from publicly available datasets. We found that RGS2 is barely detectable on the mRNA level in both bulk tissue (median 8.2. normalized mRNA reads) and single-cell data (median 0 normalized counts), but variably present on the protein level (median 34.5% positive tumor cells, moderate/strong expression in approximately 50% of samples). Interestingly, low expression of RGS2 had a negative impact on overall survival (p = 0.037) and progression-free survival (p = 0.058) on the protein level in lower FIGO stages and in the absence of residual tumor burden. A similar trend was detected on the mRNA level. Our results indicated a significant prognostic impact of RGS2 protein suppression in HGSOC. Due to diverging expression patterns of RGS2 on mRNA and protein levels, posttranslational modification of RGS2 is likely. Our findings warrant further research to unravel the functional role of RGS2 in HGSOC, especially in the light of new drug discovery.
Collapse
Affiliation(s)
- Jana Ihlow
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Nanna Monjé
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Inga Hoffmann
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Philip Bischoff
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Bruno Valentin Sinn
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Wolfgang Daniel Schmitt
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Catarina Alisa Kunze
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Sylvia Darb-Esfahani
- Institute of Pathology, Berlin-Spandau, Stadtrandstraße 555, 13589 Berlin, Germany
| | - Hagen Kulbe
- Department of Obstetrics and Gynecology with Center of Oncological Surgery, European Competence Center for Ovarian Cancer, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow-Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
- Tumorbank Ovarian Cancer Network, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow-Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Elena Ioana Braicu
- Department of Obstetrics and Gynecology with Center of Oncological Surgery, European Competence Center for Ovarian Cancer, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow-Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
- Tumorbank Ovarian Cancer Network, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow-Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Jalid Sehouli
- Department of Obstetrics and Gynecology with Center of Oncological Surgery, European Competence Center for Ovarian Cancer, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow-Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
- Tumorbank Ovarian Cancer Network, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow-Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps-University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - David Horst
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Eliane Tabea Taube
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-536-033; Fax: +49-30-450-536-900
| |
Collapse
|
102
|
Heemskerk-Gerritsen BAM, Hollestelle A, van Asperen CJ, van den Beek I, van Driel WJ, van Engelen K, Gómez Garcia EB, de Hullu JA, Koudijs MJ, Mourits MJE, Hooning MJ, Boere IA. Progression-free survival and overall survival after BRCA1/2-associated epithelial ovarian cancer: A matched cohort study. PLoS One 2022; 17:e0275015. [PMID: 36137114 PMCID: PMC9498928 DOI: 10.1371/journal.pone.0275015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/08/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction
Germline BRCA1/2-associated epithelial ovarian cancer has been associated with better progression-free survival and overall survival than sporadic epithelial ovarian cancer, but conclusive data are lacking.
Methods
We matched 389 BRCA1-associated and 123 BRCA2-associated epithelial ovarian cancer patients 1:1 to sporadic epithelial ovarian cancer patients on year of birth, year of diagnosis, and FIGO stage (< = IIA/> = IIB). Germline DNA test was performed before or after epithelial ovarian cancer diagnosis. All patients received chemotherapy. We used Cox proportional hazards models to estimate the associations between mutation status (BRCA1 or BRCA2 versus sporadic) and progression-free survival and overall survival. To investigate whether DNA testing after epithelial ovarian cancer diagnosis resulted in survival bias, we performed additional analyses limited to BRCA1/2-associated epithelial ovarian cancer patients with a DNA test result before cancer diagnosis (n = 73 BRCA1; n = 9 BRCA2) and their matched sporadic controls.
Results
The median follow-up was 4.4 years (range 0.1–30.1). During the first three years after epithelial ovarian cancer diagnosis, progression-free survival was better for BRCA1 (HR 0.88, 95% CI 0.74–1.04) and BRCA2 (HR 0.58, 95% CI 0.41–0.81) patients than for sporadic patients. Overall survival was better during the first six years after epithelial ovarian cancer for BRCA1 (HR 0.7, 95% CI 0.58–0.84) and BRCA2 (HR 0.41, 95% CI 0.29–0.59) patients. After surviving these years, survival benefits disappeared or were in favor of the sporadic patients.
Conclusion
For epithelial ovarian cancer patients who received chemotherapy, we confirmed survival benefit for BRCA1 and BRCA2 germline pathogenic variant carriers. This may indicate higher sensitivity to chemotherapy, both in first line treatment and in the recurrent setting. The observed benefit appears to be limited to a relatively short period after epithelial ovarian cancer diagnosis.
Collapse
Affiliation(s)
| | | | - Christi J. van Asperen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Irma van den Beek
- Department of Human Genetics, Amsterdam University Medical Center (University of Amsterdam), Amsterdam, the Netherlands
| | | | - Klaartje van Engelen
- Department of Clinical Genetics, Amsterdam University Medical Center (VUmc), Amsterdam, the Netherlands
| | - Encarna B. Gómez Garcia
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Joanne A. de Hullu
- Department of Obstetrics & Gynecology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marco J. Koudijs
- Department of Biomedical Genetics, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Marian J. E. Mourits
- Department of Gynecologic Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Maartje J. Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ingrid A. Boere
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
103
|
Liang YX, Zhang LL, Yang L. circANKRD17(has_circ_0007883) confers paclitaxel resistance of ovarian cancer via interacting with FUS to stabilize FOXR2. Mol Cell Biochem 2022; 478:835-850. [PMID: 36107285 DOI: 10.1007/s11010-022-04548-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 08/26/2022] [Indexed: 12/21/2022]
Abstract
Emerging numbers of endogenous circular RNAs (circRNAs) have gained much attention to serve as essential regulators in the carcinogenesis of human cancers. Unfortunately, the occurrence of paclitaxel (PTX) resistance to ovarian cancer remains to be responsible for the poor prognosis. Herein, the aim of our study is to reveal a dysregulation of a particular circRNA, circANKRD17 (has_circ_0007883), and its exact role involving in chemoresistance of ovarian cancer. Expression patterns of circANKRD17 in PTX-resistant ovarian cancer tissues and cell lines was examined using quantitative real-time PCR analysis. Role of circANKRD17 on drug resistance and cell viability was evaluated by CCK-8 assay. Colony formation was subjected to measure cell proliferation. Flow cytometry was employed to evaluate cell cycle either or cell apoptosis. Xenograft models were constructed for further in vivo confirmation. The cicrANKRD17/FUS/FOXR2 axis was demonstrated using bioinformatics analysis, RNA pull-down, as well as RNA immunoprecipitation assays. Dramatically high expressed circANKRD17 observed in ovarian cancer tissues and cells was correlated with PTX resistance, which indicated the poor prognosis. Functionally, knockdown of circANKRD17 decreased PTX resistance via inhibiting cell viability and inducing cell apoptosis. Mechanistically, circANKRD17 interacted with the RNA-binding protein, fused in sarcoma (FUS) to stabilize FOXR2. In summary, our study uncovered a novel machinery of circANKRD17/FUS/FOXR2 referring to ovarian cancer drug sensitivity and tumorigenesis, highlighting a potential strategy for circRNAs in chemoresistance.
Collapse
Affiliation(s)
- Yue-Xiu Liang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, Guangxi, People's Republic of China
| | - Lian-Ling Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, Guangxi, People's Republic of China
| | - Li Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, Guangxi, People's Republic of China.
| |
Collapse
|
104
|
Immune Tumor Microenvironment in Ovarian Cancer Ascites. Int J Mol Sci 2022; 23:ijms231810692. [PMID: 36142615 PMCID: PMC9504085 DOI: 10.3390/ijms231810692] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/26/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer (OC) has a specific type of metastasis, via transcoelomic, and most of the patients are diagnosed at advanced stages with multiple tumors spread within the peritoneal cavity. The role of Malignant Ascites (MA) is to serve as a transporter of tumor cells from the primary location to the peritoneal wall or to the surface of the peritoneal organs. MA comprise cellular components with tumor and non-tumor cells and acellular components, creating a unique microenvironment capable of modifying the tumor behavior. These microenvironment factors influence tumor cell proliferation, progression, chemoresistance, and immune evasion, suggesting that MA play an active role in OC progression. Tumor cells induce a complex immune suppression that neutralizes antitumor immunity, leading to disease progression and treatment failure, provoking a tumor-promoting environment. In this review, we will focus on the High-Grade Serous Carcinoma (HGSC) microenvironment with special attention to the tumor microenvironment immunology.
Collapse
|
105
|
Bound NT, Vandenberg CJ, Kartikasari AER, Plebanski M, Scott CL. Improving PARP inhibitor efficacy in high-grade serous ovarian carcinoma: A focus on the immune system. Front Genet 2022; 13:886170. [PMID: 36159999 PMCID: PMC9505691 DOI: 10.3389/fgene.2022.886170] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/05/2022] [Indexed: 12/03/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is a genomically unstable malignancy responsible for over 70% of all deaths due to ovarian cancer. With roughly 50% of all HGSOC harboring defects in the homologous recombination (HR) DNA repair pathway (e.g., BRCA1/2 mutations), the introduction of poly ADP-ribose polymerase inhibitors (PARPi) has dramatically improved outcomes for women with HR defective HGSOC. By blocking the repair of single-stranded DNA damage in cancer cells already lacking high-fidelity HR pathways, PARPi causes the accumulation of double-stranded DNA breaks, leading to cell death. Thus, this synthetic lethality results in PARPi selectively targeting cancer cells, resulting in impressive efficacy. Despite this, resistance to PARPi commonly develops through diverse mechanisms, such as the acquisition of secondary BRCA1/2 mutations. Perhaps less well documented is that PARPi can impact both the tumour microenvironment and the immune response, through upregulation of the stimulator of interferon genes (STING) pathway, upregulation of immune checkpoints such as PD-L1, and by stimulating the production of pro-inflammatory cytokines. Whilst targeted immunotherapies have not yet found their place in the clinic for HGSOC, the evidence above, as well as ongoing studies exploring the synergistic effects of PARPi with immune agents, including immune checkpoint inhibitors, suggests potential for targeting the immune response in HGSOC. Additionally, combining PARPi with epigenetic-modulating drugs may improve PARPi efficacy, by inducing a BRCA-defective phenotype to sensitise resistant cancer cells to PARPi. Finally, invigorating an immune response during PARPi therapy may engage anti-cancer immune responses that potentiate efficacy and mitigate the development of PARPi resistance. Here, we will review the emerging PARPi literature with a focus on PARPi effects on the immune response in HGSOC, as well as the potential of epigenetic combination therapies. We highlight the potential of transforming HGSOC from a lethal to a chronic disease and increasing the likelihood of cure.
Collapse
Affiliation(s)
- Nirashaa T. Bound
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Cassandra J. Vandenberg
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Apriliana E. R. Kartikasari
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Magdalena Plebanski
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Clare L. Scott
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Royal Women’s Hospital, Parkville, VIC, Australia
| |
Collapse
|
106
|
Zalfa F, Perrone MG, Ferorelli S, Laera L, Pierri CL, Tolomeo A, Dimiccoli V, Perrone G, De Grassi A, Scilimati A. Genome-Wide Identification and Validation of Gene Expression Biomarkers in the Diagnosis of Ovarian Serous Cystadenocarcinoma. Cancers (Basel) 2022; 14:cancers14153764. [PMID: 35954427 PMCID: PMC9367275 DOI: 10.3390/cancers14153764] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Despite ovarian serous adenocarcinoma (OSCA) is a high-incidence type of cancer, limited molecular screening methods are available and the diagnosis mostly occurs at a late stage. The aim of this study is screening the potential of gene expression for identifying OSCA-specific molecular biomarkers for improving diagnosis. A genome-wide survey was performed on high-throughput RNA-sequencing experiments on hundreds ovarian cancer samples and healthy ovarian tissues, providing a number of putative OSCA biomarkers, which were then validated on an independent sample set and using a different RNA-quantification technology. Combinations of gene expression biomarkers were identified, which showed high accuracy in discriminating OSCA tissues from the normal counterpart and from other tumor types. The detected biomarkers can improve the molecular diagnosis of OSCA on tissue samples and are, in principle, translatable to the analysis of liquid biopsies. Abstract Ovarian cancer is the second most prevalent gynecologic malignancy, and ovarian serous cystadenocarcinoma (OSCA) is the most common and lethal subtype of ovarian cancer. Current screening methods have strong limits on early detection, and the majority of OSCA patients relapse. In this work, we developed and cross-validated a method for detecting gene expression biomarkers able to discriminate OSCA tissues from healthy ovarian tissues and other cancer types with high accuracy. A preliminary ranking-based approach was applied, resulting in a panel of 41 over-expressed genes in OSCA. The RNA quantity gene expression of the 41 selected genes was then cross-validated by using NanoString nCounter technology. Moreover, we showed that the RNA quantity of eight genes (ADGRG1, EPCAM, ESRP1, MAL2, MYH14, PRSS8, ST14 and WFDC2) discriminates each OSCA sample from each healthy sample in our data set with sensitivity of 100% and specificity of 100%. For the other three genes (MUC16, PAX8 and SOX17) in combination, their RNA quantity may distinguish OSCA from other 29 tumor types.
Collapse
Affiliation(s)
- Francesca Zalfa
- Predictive Molecular Diagnostic Unit, Pathology Department, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
- Microscopic and Ultrastructural Anatomy Unit, CIR, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Maria Grazia Perrone
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.G.P.); (S.F.)
| | - Savina Ferorelli
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.G.P.); (S.F.)
| | - Luna Laera
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari “Aldo Moro”, 70125 Bari, Italy; (L.L.); (C.L.P.)
| | - Ciro Leonardo Pierri
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari “Aldo Moro”, 70125 Bari, Italy; (L.L.); (C.L.P.)
| | - Anna Tolomeo
- Department of ITELPHARMA, ITEL Telecomunicazioni S.R.L., 70037 Ruvo di Puglia, Italy; (A.T.); (V.D.)
| | - Vincenzo Dimiccoli
- Department of ITELPHARMA, ITEL Telecomunicazioni S.R.L., 70037 Ruvo di Puglia, Italy; (A.T.); (V.D.)
| | - Giuseppe Perrone
- Pathology Department, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
- Pathology Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Anna De Grassi
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari “Aldo Moro”, 70125 Bari, Italy; (L.L.); (C.L.P.)
- Correspondence: (A.D.G.); (A.S.)
| | - Antonio Scilimati
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.G.P.); (S.F.)
- Correspondence: (A.D.G.); (A.S.)
| |
Collapse
|
107
|
Zırh EB, Kapaklı ET, Dolgun A, Usubütün A, Zeybek ND. The expression of BMP, integrin, ZEB2 in ovarian high-grade serous carcinoma in relation with lymph node metastasis. Growth Factors 2022; 40:153-162. [PMID: 35867635 DOI: 10.1080/08977194.2022.2099849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Ovarian cancer (OC) is clinically important because it is diagnosed late and has metastasis when it is diagnosed. Mortality risk increases 2.75 times in the presence of lymph node (LN) metastasis. During metastasis, many molecules including BMPs originated from stroma, and tumor cells participate through transcription factors and integrins for cytoskeleton regulation during cell migration. We hypothesized an inverse correlation between BMP2 and BMP7 along with changes in ZEB2, and integrin α5β1 in high-grade OCs in relation to LN metastasis. The BMP2 immunoreactivity was strong along with strong ZEB2 and weak integrins' immunoreactivity in samples with LN metastasis. Strong immunoreactivity of BMP7 was accompanied by strong immunoreactivity of integrins in the samples without LN metastasis. Study results showed BMP2's strong positive immunoreactivity and weak BMP7 immunoreactivity in tumor cells with a significantly weak inverse correlation. This inverse correlation should be considered as both BMPs have different effects in the window of cancer progression and invasion.
Collapse
Affiliation(s)
- Elham Bahador Zırh
- Department of Histology and Embryology, Faculty of Medicine, TOBB Economy and Technology University, Ankara, Turkey
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Elif Taşar Kapaklı
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Anıl Dolgun
- Department of Mathematical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Alp Usubütün
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Naciye Dilara Zeybek
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
108
|
Yu R, Hu Y, Zhang S, Li X, Tang M, Yang M, Wu X, Li Z, Liao X, Xu Y, Li M, Chen S, Qian W, Gong LY, Song L, Li J. LncRNA CTBP1-DT-encoded microprotein DDUP sustains DNA damage response signalling to trigger dual DNA repair mechanisms. Nucleic Acids Res 2022; 50:8060-8079. [PMID: 35849344 PMCID: PMC9371908 DOI: 10.1093/nar/gkac611] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/21/2022] [Accepted: 07/01/2022] [Indexed: 11/14/2022] Open
Abstract
Sustaining DNA damage response (DDR) signalling via retention of DDR factors at damaged sites is important for transmitting damage-sensing and repair signals. Herein, we found that DNA damage provoked the association of ribosomes with IRES region in lncRNA CTBP1-DT, which overcame the negative effect of upstream open reading frames (uORFs), and elicited the novel microprotein DNA damage-upregulated protein (DDUP) translation via a cap-independent translation mechanism. Activated ATR kinase-mediated phosphorylation of DDUP induced a drastic 'dense-to-loose' conformational change, which sustained the RAD18/RAD51C and RAD18/PCNA complex at damaged sites and initiated RAD51C-mediated homologous recombination and PCNA-mediated post-replication repair mechanisms. Importantly, treatment with ATR inhibitor abolished the effect of DDUP on chromatin retention of RAD51C and PCNA, thereby leading to hypersensitivity of cancer cells to DNA-damaging chemotherapeutics. Taken together, our results uncover a plausible mechanism underlying the DDR sustaining and might represent an attractive therapeutic strategy in improvement of DNA damage-based anticancer therapies.
Collapse
Affiliation(s)
- Ruyuan Yu
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Yameng Hu
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Shuxia Zhang
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Xincheng Li
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Miaoling Tang
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Meisongzhu Yang
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Xingui Wu
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Ziwen Li
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Xinyi Liao
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Yingru Xu
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Man Li
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Suwen Chen
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Wanying Qian
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Li-Yun Gong
- Guangdong Provincial Key Laboratory for Genome Stability and Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen University, China
| | - Libing Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, China
| | - Jun Li
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| |
Collapse
|
109
|
Revythis A, Limbu A, Mikropoulos C, Ghose A, Sanchez E, Sheriff M, Boussios S. Recent Insights into PARP and Immuno-Checkpoint Inhibitors in Epithelial Ovarian Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:8577. [PMID: 35886427 PMCID: PMC9317199 DOI: 10.3390/ijerph19148577] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/06/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023]
Abstract
Ovarian cancer is one of the most common gynecologic cancers and has the highest mortality rate of any other cancer of the female reproductive system. Epithelial ovarian cancer (EOC) accounts for approximately 90% of all ovarian malignancies. The standard therapeutic strategy includes cytoreductive surgery accompanied by pre- or postoperative platinum-based chemotherapy. Nevertheless, up to 80% of the patients relapse within the following 12-18 months from the completion of the treatment and then receive first-line chemotherapy depending on platinum sensitivity. Mutations in BRCA1/2 genes are the most significant molecular aberrations in EOC and serve as prognostic and predictive biomarkers. Poly ADP-ribose polymerase (PARP) inhibitors exploit defects in the DNA repair pathway through synthetic lethality. They have also been shown to trap PARP1 and PARP2 on DNA, leading to PARP-DNA complexes. Olaparib, rucaparib, and niraparib have all obtained Food and Drug Administration (FDA) and/or the European Medicine Agency (EMA) approval for the treatment of EOC in different settings. Immune checkpoint inhibitors (ICI) have improved the survival of several cancers and are under evaluation in EOC. However, despite the success of immunotherapy in other malignancies, the use of antibodies inhibiting the immune checkpoint programmed cell death (PD-1) or its ligand (PD-L1) obtained modest results in EOC so far, with median response rates of up to 10%. As such, ICI have not yet been approved for the treatment of EOC. We herein provided a comprehensive insight into the most recent progress in synthetic lethality PARP inhibitors, along with the mechanisms of resistance. We also summarised data regarding the role of immune checkpoint inhibitors, the use of vaccination therapy, and adoptive immunotherapy in treating epithelial ovarian cancer.
Collapse
Affiliation(s)
- Antonios Revythis
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
| | - Anu Limbu
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
| | - Christos Mikropoulos
- St. Lukes Cancer Centre, Royal Surrey County Hospital, Egerton Rd., Guildford GU2 7XX, Surrey, UK;
| | - Aruni Ghose
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
- Department of Medical Oncology, Barts Cancer Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London KT1 2EE, UK
- Department of Medical Oncology, Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, London KT1 2EE, UK
- Centre for Education, Faculty of Life Sciences and Medicine, King’s College London, London SE5 9NU, UK
| | - Elisabet Sanchez
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
| | - Matin Sheriff
- Department of Urology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK;
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK; (A.R.); (A.L.); (A.G.); (E.S.)
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
| |
Collapse
|
110
|
Dhiman P, Bapsy P, Patil C, Raghupathi R. Is Optimal Cytoreduction Post Neoadjuvant Chemotherapy the Only Prognostic Factor in Advanced Ovarian Cancer? South Asian J Cancer 2022. [DOI: 10.1055/s-0042-1754441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Background Epithelial ovarian cancer (EOC) is one of the leading causes of cancer-related death in women. Approximately 70% of patients with EOC are diagnosed in advanced stage [The International Federation of Gynecology and Obstetrics(FIGO stage III and IV)] with an expected 5-year survival rate of 30%. Numerous studies have shown that survival with neoadjuvant chemotherapy (NACT) followed by interval debulking surgery (IDS) is noninferior to primary debulking surgery followed by chemotherapy.
Materials and Methods In this retroprospective observational study, 50 patients with advanced ovarian cancer, diagnosed from January 2012 to January 2015, were included and followed-up till January 2017. Correlation of NACT with patient profile, CA125 levels, clinicopathologic parameters, progression-free survival (PFS), and treatment response was studied. Statistical analysis was performed using log-rank test and Kaplan-Meir survival plots.
Results The extent of cytoreduction significantly correlated with PFS. The PFS was maximum in patients who had optimal cytoreduction (19 months) and 10 months in patients with suboptimal cytoreduction with p-value < 0.05. The survival was not significantly correlated with other parameters such as age, stage, preoperative CA125 levels, and ascites.
Conclusions The extent of cytoreduction following NACT in this study was associated with statistically significant PFS advantage in patients who were able to undergo optimal cytoreduction, but not significantly correlated to other factors such as age, stage, preoperative CA125 levels, and ascites. NACT followed by interval cytoreduction is an important modality affecting survival in advanced EOC. Further studies and longer follow-up are needed to demonstrate survival advantage over standard treatment.
Collapse
Affiliation(s)
- Pravesh Dhiman
- Medical Oncology, Department of RT and Oncology, IGMC, Shimla, Himachal Pradesh, India
| | - P.P. Bapsy
- Medical Oncology, Department of Medical Oncology, Apollo Hospitals, Bangalore, Karnataka, India
| | - C.N. Patil
- Medical Oncology, Department of Medical Oncology, Apollo Hospitals, Bangalore, Karnataka, India
| | - Renu Raghupathi
- Medical Oncology, Department of Medical Oncology, Apollo Hospitals, Bangalore, Karnataka, India
| |
Collapse
|
111
|
Extraterrestrial Gynecology: Could Spaceflight Increase the Risk of Developing Cancer in Female Astronauts? An Updated Review. Int J Mol Sci 2022; 23:ijms23137465. [PMID: 35806469 PMCID: PMC9267413 DOI: 10.3390/ijms23137465] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
Outer space is an extremely hostile environment for human life, with ionizing radiation from galactic cosmic rays and microgravity posing the most significant hazards to the health of astronauts. Spaceflight has also been shown to have an impact on established cancer hallmarks, possibly increasing carcinogenic risk. Terrestrially, women have a higher incidence of radiation-induced cancers, largely driven by lung, thyroid, breast, and ovarian cancers, and therefore, historically, they have been permitted to spend significantly less time in space than men. In the present review, we focus on the effects of microgravity and radiation on the female reproductive system, particularly gynecological cancer. The aim is to provide a summary of the research that has been carried out related to the risk of gynecological cancer, highlighting what further studies are needed to pave the way for safer exploration class missions, as well as postflight screening and management of women astronauts following long-duration spaceflight.
Collapse
|
112
|
Cotler MJ, Ramadi KB, Hou X, Christodoulopoulos E, Ahn S, Bashyam A, Ding H, Larson M, Oberg AL, Whittaker C, Jonas O, Kaufmann SH, Weroha SJ, Cima MJ. Machine-learning aided in situ drug sensitivity screening predicts treatment outcomes in ovarian PDX tumors. Transl Oncol 2022; 21:101427. [PMID: 35472731 PMCID: PMC9136609 DOI: 10.1016/j.tranon.2022.101427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/28/2022] [Accepted: 04/10/2022] [Indexed: 12/14/2022] Open
Abstract
Long-term treatment outcomes for patients with high grade ovarian cancers have not changed despite innovations in therapies. There is no recommended assay for predicting patient response to second-line therapy, thus clinicians must make treatment decisions based on each individual patient. Patient-derived xenograft (PDX) tumors have been shown to predict drug sensitivity in ovarian cancer patients, but the time frame for intraperitoneal (IP) tumor generation, expansion, and drug screening is beyond that for tumor recurrence and platinum resistance to occur, thus results do not have clinical utility. We describe a drug sensitivity screening assay using a drug delivery microdevice implanted for 24 h in subcutaneous (SQ) ovarian PDX tumors to predict treatment outcomes in matched IP PDX tumors in a clinically relevant time frame. The SQ tumor response to local microdose drug exposure was found to be predictive of the growth of matched IP tumors after multi-week systemic therapy using significantly fewer animals (10 SQ vs 206 IP). Multiplexed immunofluorescence image analysis of phenotypic tumor response combined with a machine learning classifier could predict IP treatment outcomes against three second-line cytotoxic therapies with an average AUC of 0.91.
Collapse
Affiliation(s)
- Max J Cotler
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard-MIT Program in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Khalil B Ramadi
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard-MIT Program in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xiaonan Hou
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Elena Christodoulopoulos
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sebastian Ahn
- Department of Radiology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ashvin Bashyam
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Huiming Ding
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Melissa Larson
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Charles Whittaker
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Oliver Jonas
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Radiology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Scott H Kaufmann
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - S John Weroha
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael J Cima
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
113
|
Dhiman P, Bapsy P, Patil C, Raghupathi R. Is Optimal Cytoreduction Post Neoadjuvant Chemotherapy the Only Prognostic Factor in Advanced Ovarian Cancer? South Asian J Cancer 2022; 11:207-212. [PMID: 36588609 PMCID: PMC10497344 DOI: 10.1055/s-0042-1755291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Pravesh Dhiman, DNB (Medical Oncology).Background Epithelial ovarian cancer (EOC) is one of the leading causes of cancer-related death in women. Approximately 70% of patients with EOC are diagnosed in advanced stage [The International Federation of Gynecology and Obstetrics(FIGO stage III and IV)] with an expected 5-year survival rate of 30%. Numerous studies have shown that survival with neoadjuvant chemotherapy (NACT) followed by interval debulking surgery (IDS) is noninferior to primary debulking surgery followed by chemotherapy. Materials and Methods In this retroprospective observational study, 50 patients with advanced ovarian cancer, diagnosed from January 2012 to January 2015, were included and followed-up till January 2017. Correlation of NACT with patient profile, CA125 levels, clinicopathologic parameters, progression-free survival (PFS), and treatment response was studied. Statistical analysis was performed using log-rank test and Kaplan-Meir survival plots. Results The extent of cytoreduction significantly correlated with PFS. The PFS was maximum in patients who had optimal cytoreduction (19 months) and 10 months in patients with suboptimal cytoreduction with p -value < 0.05. The survival was not significantly correlated with other parameters such as age, stage, preoperative CA125 levels, and ascites. Conclusions The extent of cytoreduction following NACT in this study was associated with statistically significant PFS advantage in patients who were able to undergo optimal cytoreduction, but not significantly correlated to other factors such as age, stage, preoperative CA125 levels, and ascites. NACT followed by interval cytoreduction is an important modality affecting survival in advanced EOC. Further studies and longer follow-up are needed to demonstrate survival advantage over standard treatment.
Collapse
Affiliation(s)
- Pravesh Dhiman
- Medical Oncology, Department of RT and Oncology, IGMC, Shimla, Himachal Pradesh, India
| | - P.P. Bapsy
- Medical Oncology, Department of Medical Oncology, Apollo Hospitals, Bangalore, Karnataka, India
| | - C.N. Patil
- Medical Oncology, Department of Medical Oncology, Apollo Hospitals, Bangalore, Karnataka, India
| | - Renu Raghupathi
- Medical Oncology, Department of Medical Oncology, Apollo Hospitals, Bangalore, Karnataka, India
| |
Collapse
|
114
|
Shah S, Cheung A, Kutka M, Sheriff M, Boussios S. Epithelial Ovarian Cancer: Providing Evidence of Predisposition Genes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19138113. [PMID: 35805770 PMCID: PMC9265838 DOI: 10.3390/ijerph19138113] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/19/2022]
Abstract
Epithelial ovarian cancer (EOC) is one of the cancers most influenced by hereditary factors. A fourth to a fifth of unselected EOC patients carry pathogenic variants (PVs) in a number of genes, the majority of which encode for proteins involved in DNA mismatch repair (MMR) pathways. PVs in BRCA1 and BRCA2 genes are responsible for a substantial fraction of hereditary EOC. In addition, PV genes involved in the MMR pathway account for 10–15% of hereditary EOC. The identification of women with homologous recombination (HR)-deficient EOCs has significant clinical implications, concerning chemotherapy regimen planning and development as well as the use of targeted therapies such as poly(ADP-ribose) polymerase (PARP) inhibitors. With several genes involved, the complexity of genetic testing increases. In this context, next-generation sequencing (NGS) allows testing for multiple genes simultaneously with a rapid turnaround time. In this review, we discuss the EOC risk assessment in the era of NGS.
Collapse
Affiliation(s)
- Sidrah Shah
- Department of Palliative Care, Guy’s and St Thomas’ Hospital, London SE1 9RT, UK;
| | - Alison Cheung
- Department of Hematology/Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Kent, Gillingham ME7 5NY, UK; (A.C.); (M.K.)
| | - Mikolaj Kutka
- Department of Hematology/Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Kent, Gillingham ME7 5NY, UK; (A.C.); (M.K.)
| | - Matin Sheriff
- Department of Urology, Medway NHS Foundation Trust, Windmill Road, Kent, Gillingham ME7 5NY, UK;
| | - Stergios Boussios
- Department of Palliative Care, Guy’s and St Thomas’ Hospital, London SE1 9RT, UK;
- King’s College London, Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
- Correspondence: or or
| |
Collapse
|
115
|
Zhu Y, Yang L, Chen Y. Anhuienoside C inhibits human ovarian cancer cell growth by inducing apoptosis, suppression of cell migration and invasion, and targeting PI3K/AKT/mTOR signaling pathway. Mol Cell Biochem 2022; 477:1887-1892. [PMID: 35348981 DOI: 10.1007/s11010-022-04406-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/02/2022] [Indexed: 11/26/2022]
Abstract
The present study was initiated to examine the anticancer effects of Anhuienoside C (AC) against ovarian cancer and postulates the possible molecular mechanism of its action. 3-[4,5-Dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide assay was implemented for determination of the effects of AC on cell viability of the ovarian cancer OVACAR-3 cell line. To study cellular morphology, phase contrast microscopy was performed. Apoptosis was examined via acridine orange/ethidium bromide used staining assays. Flow cytometry was used to check the different phases of the cell cycle. Cell migration and invasion assays were performed via transwell chamber assay. The effects of AC on expression of phosphoinositide 3-kinases (PI3K), protein kinase B (AKT), and mammalian target of rapamycin (mTOR) protein in ovarian cell were assessed using western blotting assay. The results indicated that the cell proliferation rate lowered in AC-treated OVACAR-3 cells as compared to the untreated controls in a dose-dependent manner. Cell morphology changed substantially by the exposure to AC and remained dose dependent. These morphological changes were indicative of apoptotic cell death. Apoptosis analysis showed dose-dependent increase of apoptosis. The cell migration and invasion of OVACAR-3 cells was reduced to a minimum by AC in a dose-dependent manner. Finally, western blotting assay showed blocking of PI3K/AKT/mTOR signaling pathway with increasing AC doses. Taking all together, AC is a potential ovarian cancer inhibitor. It induces its anti-ovarian cancer effects via induction of apoptosis, delaying cell migration and invasion, and blocking PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Obstetrics and Gynecology, Yancheng No.1 People Hospital, Yancheng, 215000, Jiangsu, China
| | - Lijuan Yang
- Department of Obstetrics and Gynecology, Yancheng No.1 People Hospital, Yancheng, 215000, Jiangsu, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, 215100, Jiangsu, China.
| |
Collapse
|
116
|
Hu J, Wang Z, Zuo R, Zheng C, Lu B, Cheng X, Lu W, Zhao C, Liu P, Lu Y. Development of survival predictors for high-grade serous ovarian cancer based on stable radiomic features from computed tomography images. iScience 2022; 25:104628. [PMID: 35800777 PMCID: PMC9254345 DOI: 10.1016/j.isci.2022.104628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/03/2022] [Accepted: 06/13/2022] [Indexed: 12/04/2022] Open
Abstract
Less than 35% of advanced patients with high-grade serous ovarian cancer (HGSOC) survive for 5 years after diagnosis. Here, we developed radiomics-based models to predict HGSOC clinical outcomes using preoperative contrast-enhanced computed tomography (CECT) images. 891 radiomics features were extracted between primary, metastatic, or lymphatic lesions from preoperative venous phase CECT images of 217 patients with HGSOC. A heuristic method, Frequency Appearance in Multiple Univariate preScreening (FAMUS), was proposed to identify stable and task-relevant radiomic features. Using FAMUS, we constructed predictive models of overall survival and disease-free survival in patients with HGSOC based on these stable radiomic features. According to their CT images, patients with HGSOC can be accurately stratified into high-risk or low-risk groups for cancer-related death within 2-6 years or for likely recurrence within 1-5 years. These radiomic models provide convincing and reliable non-invasive markers for individualized prognostic evaluation and clinical decision-making for patients with HGSOC. Frequency Appearance in Multiple Univariate preScreening (FAMUS) identifies stable and task-relevant radiomic features from computed tomography (CT) images Radiomics-based signatures are highly predictive of the clinical outcome of high-grade serous ovarian cancer (HGSOC) FAMUS improves the prognostic performance of radiomics-based prediction models Developed radiomic models can help clinicians tailor treatment plans for HGSOC
Collapse
|
117
|
Fibroblast growth factor signalling influences homologous recombination-mediated DNA damage repair to promote drug resistance in ovarian cancer. Br J Cancer 2022; 127:1340-1351. [PMID: 35778553 PMCID: PMC9519926 DOI: 10.1038/s41416-022-01899-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Ovarian cancer patients frequently develop chemotherapy resistance, limiting treatment options. We have previously shown that individuality in fibroblast growth factor 1 (FGF1) expression influences survival and chemotherapy response. METHODS We used MTT assays to assess chemosensitivity to cisplatin and carboplatin following shRNA-mediated knockdown or heterologous over-expression of FGF1 (quantified by qRT-PCR and immunoblot analysis), and in combination with the FGFR inhibitors AZD4547 and SU5402, the ATM inhibitor KU55933 and DNA-PK inhibitor NU7026. Immunofluorescence microscopy was used to quantify the FGF1-dependent timecourse of replication protein A (RPA) and γH2AX foci formation. RESULTS Pharmacological inhibition of FGF signalling reversed drug resistance in immortalised cell lines and in primary cell lines from drug-resistant ovarian cancer patients, while FGF1 over-expression induced resistance. Ataxia telangiectasia mutated (ATM) phosphorylation, but not DNA adduct formation was FGF1 dependent, following cisplatin or carboplatin challenge. Combining platinum drugs with the ATM inhibitor KU55933, but not with the DNA-PK inhibitor NU7026 re-sensitised resistant cells. FGF1 expression influenced the timecourse of damage-induced RPA and γH2AX nuclear foci formation. CONCLUSION Drug resistance arises from FGF1-mediated differential activation of high-fidelity homologous recombination DNA damage repair. FGFR and ATM inhibitors reverse platinum drug resistance, highlighting novel combination chemotherapy approaches for future clinical trial evaluation.
Collapse
|
118
|
Liu J, Liu L, Antwi PA, Luo Y, Liang F. Identification and Validation of the Diagnostic Characteristic Genes of Ovarian Cancer by Bioinformatics and Machine Learning. Front Genet 2022; 13:858466. [PMID: 35719392 PMCID: PMC9198487 DOI: 10.3389/fgene.2022.858466] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Ovarian cancer (OC) has a high mortality rate and poses a severe threat to women’s health. However, abnormal gene expression underlying the tumorigenesis of OC has not been fully understood. This study aims to identify diagnostic characteristic genes involved in OC by bioinformatics and machine learning. Methods: We utilized five datasets retrieved from the Gene Expression Omnibus (GEO) database, The Cancer Genome Atlas (TCGA) database, and the Genotype-Tissue Expression (GTEx) Project database. GSE12470 and GSE18520 were combined as the training set, and GSE27651 was used as the validation set A. Also, we combined the TCGA database and GTEx database as validation set B. First, in the training set, differentially expressed genes (DEGs) between OC and non-ovarian cancer tissues (nOC) were identified. Next, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Disease Ontology (DO) enrichment analysis, and Gene Set Enrichment Analysis (GSEA) were performed for functional enrichment analysis of these DEGs. Then, two machine learning algorithms, Least Absolute Shrinkage and Selector Operation (LASSO) and Support Vector Machine-Recursive Feature Elimination (SVM-RFE), were used to get the diagnostic genes. Subsequently, the obtained diagnostic-related DEGs were validated in the validation sets. Then, we used the computational approach (CIBERSORT) to analyze the association between immune cell infiltration and DEGs. Finally, we analyzed the prognostic role of several genes on the KM-plotter website and used the human protein atlas (HPA) online database to analyze the expression of these genes at the protein level. Results: 590 DEGs were identified, including 276 upregulated and 314 downregulated DEGs.The Enrichment analysis results indicated the DEGs were mainly involved in the nuclear division, cell cycle, and IL−17 signaling pathway. Besides, DEGs were also closely related to immune cell infiltration. Finally, we found that BUB1, FOLR1, and PSAT1 have prognostic roles and the protein-level expression of these six genes SFPR1, PSAT1, PDE8B, INAVA and TMEM139 in OC tissue and nOC tissue was consistent with our analysis. Conclusions: We screened nine diagnostic characteristic genes of OC, including SFRP1, PSAT1, BUB1B, FOLR1, ABCB1, PDE8B, INAVA, BUB1, TMEM139. Combining these genes may be useful for OC diagnosis and evaluating immune cell infiltration.
Collapse
Affiliation(s)
- Jinya Liu
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Leping Liu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Paul Akwasi Antwi
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yanwei Luo
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Fang Liang
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
119
|
Hollis RL, Croy I, Churchman M, Bartos C, Rye T, Gourley C, Herrington CS. Ovarian carcinosarcoma is a distinct form of ovarian cancer with poorer survival compared to tubo-ovarian high-grade serous carcinoma. Br J Cancer 2022; 127:1034-1042. [PMID: 35715633 PMCID: PMC9470739 DOI: 10.1038/s41416-022-01874-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 11/09/2022] Open
Abstract
Background Ovarian carcinosarcoma (OCS) is an uncommon, biphasic and highly aggressive ovarian cancer type, which has received relatively little research attention. Methods We curated the largest pathologically confirmed OCS cohort to date, performing detailed histopathological characterisation, analysis of features associated with survival and comparison against high-grade serous ovarian carcinoma (HGSOC). Results Eighty-two OCS patients were identified; overall survival was poor (median 12.7 months). In all, 79% demonstrated epithelial components of high-grade serous (HGS) type, while 21% were endometrioid. Heterologous elements were common (chondrosarcoma in 32%, rhabdomyosarcoma in 21%, liposarcoma in 2%); chondrosarcoma was more frequent in OCS with endometrioid carcinomatous components. Earlier stage, complete resection and platinum-containing adjuvant chemotherapy were associated with prolonged survival; however, risk of relapse and mortality was high across all patient groups. Histological subclassification did not identify subgroups with distinct survival. Compared to HGSOC, OCS patients were older (P < 0.0001), more likely to be FIGO stage I (P = 0.025), demonstrated lower chemotherapy response rate (P = 0.001) and had significantly poorer survival (P < 0.0001). Conclusion OCS represents a distinct, highly lethal form of ovarian cancer for which new treatment strategies are urgently needed. Histological subclassification does not identify patient subgroups with distinct survival. Aggressive adjuvant chemotherapy should be considered for all cases, including those with early-stage disease.
Collapse
Affiliation(s)
- Robert L Hollis
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK.
| | - Ian Croy
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | - Michael Churchman
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | - Clare Bartos
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | - Tzyvia Rye
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | - Charlie Gourley
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | - C Simon Herrington
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
120
|
Sa JK, Kim J, Kang S, Kim SW, Song T, Shim SH, Choi MC, No JH, Song JY, Kim D, Kim YM, Kim JH, Lee JW. Somatic genomic landscape of East Asian epithelial ovarian carcinoma and its clinical implications from prospective clinical sequencing; A Korean Gynecologic Oncology Group study (KGOG 3047). Int J Cancer 2022; 151:1086-1097. [PMID: 35666535 DOI: 10.1002/ijc.34150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/27/2022] [Accepted: 05/17/2022] [Indexed: 12/24/2022]
Abstract
Through the wide adaptation of next-generation sequencing (NGS) technology within clinical practice, molecular profiling of the tumor has been the principal component of personalized treatment. In this study, we have generated a large collection of cancer genomes on East Asian epithelial ovarian carcinoma (EOC) patients and demonstrate the feasibility and utility of NGS platforms to explore the dynamic interrelations of major cancer driver alterations and their impacts on clinical prognosis and management. A total of 652 EOC patients have undergone clinical NGS panels to determine the prevalence of germline and somatic mutations. Notably, TP53 was the most frequently altered event (73%), followed by both BRCA1 and BRCA2 (22% each) and MYC (19%) through pan-EOC analysis. When analyzed based on individual histopathological levels, TP53 mutation was highly dominant in high-grade serous and mucinous histology, whereas mutations in PIK3CA and ARID1A were mostly observed in clear cell carcinoma, and KRAS, BRAF, and CDKN2A mutations were enriched in endometrioid, low-grade serous, and mucinous tumors, respectively. The network-based probabilistic model showed significant co-occurrences of TP53 with BRCA1 and ALK with BRCA2, NOTCH1, and ROS1, whereas mutual exclusivity of TP53 with KRAS and PIK3CA was evident. Furthermore, we utilized machine-learning algorithms to identify molecular correlates that conferred increased sensitivity to platinum and olaparib treatments including somatic mutations in BRCA1, ATM, and MYC. Conversely, patients with ALK mutation were considerably resistant to both treatment modalities. Collectively, our results demonstrate the clinical feasibility of prospective genetic sequencing to facilitate personalized treatment opportunities for patients with EOC.
Collapse
Affiliation(s)
- Jason K Sa
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jihye Kim
- Departments of Obstetrics and Gynecology, Chung-ang University Gwang-myeong Hospital, Gwang-myeong, Republic of Korea
| | - Sokbom Kang
- Gynecologic Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Sang Wun Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Women's Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Taejong Song
- Department of Obstetrics and Gynecology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seung-Hyuk Shim
- Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Min Chul Choi
- Comprehensive Gynecologic Cancer Center, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jae Hong No
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jae-Yun Song
- Department of Obstetrics and Gynecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Deokhoon Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong-Man Kim
- Department of Obstetrics and Gynecology, College of Medicine, University of Ulsan, Asan Medical Center, Seoul, Korea
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong-Won Lee
- Departments of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
121
|
Chemoresistant Cancer Cell Lines Are Characterized by Migratory, Amino Acid Metabolism, Protein Catabolism and IFN1 Signalling Perturbations. Cancers (Basel) 2022; 14:cancers14112763. [PMID: 35681748 PMCID: PMC9179525 DOI: 10.3390/cancers14112763] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary While chemoresistance remains a major barrier to improving the outcomes for patients with ovarian cancer, the molecular features, and associated biological functions, which underpin chemoresistance in ovarian cancer remain poorly understood. In this study we aimed to provide insight into the proteins and metabolites, and their associated biological pathways, which play a role in conferring chemoresistance to ovarian cancer. Through mass spectrometry analysis comparing the proteome and metabolome of chemosensitive vs chemoresistant ovarian cancer cell lines we revealed numerous perturbations in signalling and metabolic pathways in chemoresistant cells. Further comparison to primary cells taken from patients with chemoresistant or chemosensitive disease identified a shared dysregulation in cytokine and type 1 interferon signalling. Our research sets the foundation for a deeper understanding of the proteomic and metabolomic features of chemoresistance and identifies type 1 interferon signalling as a common feature of chemoresistance. Abstract Chemoresistance remains the major barrier to effective ovarian cancer treatment. The molecular features and associated biological functions of this phenotype remain poorly understood. We developed carboplatin-resistant cell line models using OVCAR5 and CaOV3 cell lines with the aim of identifying chemoresistance-specific molecular features. Chemotaxis and CAM invasion assays revealed enhanced migratory and invasive potential in OVCAR5-resistant, compared to parental cell lines. Mass spectrometry analysis was used to analyse the metabolome and proteome of these cell lines, and was able to separate these populations based on their molecular features. It revealed signalling and metabolic perturbations in the chemoresistant cell lines. A comparison with the proteome of patient-derived primary ovarian cancer cells grown in culture showed a shared dysregulation of cytokine and type 1 interferon signalling, potentially revealing a common molecular feature of chemoresistance. A comprehensive analysis of a larger patient cohort, including advanced in vitro and in vivo models, promises to assist with better understanding the molecular mechanisms of chemoresistance and the associated enhancement of migration and invasion.
Collapse
|
122
|
Chiappetta G, Gamberi T, Faienza F, Limaj X, Rizza S, Messori L, Filomeni G, Modesti A, Vinh J. Redox proteome analysis of auranofin exposed ovarian cancer cells (A2780). Redox Biol 2022; 52:102294. [PMID: 35358852 PMCID: PMC8966199 DOI: 10.1016/j.redox.2022.102294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023] Open
Abstract
The effects of Auranofin (AF) on protein expression and protein oxidation in A2780 cancer cells were investigated through a strategy based on simultaneous expression proteomics and redox proteomics determinations. Bioinformatics analysis of the proteomics data supports the view that the most critical cellular changes elicited by AF treatment consist of thioredoxin reductase inhibition, alteration of the cell redox state, impairment of the mitochondrial functions, metabolic changes associated with conversion to a glycolytic phenotype, induction of ER stress. The occurrence of the above cellular changes was extensively validated by performing direct biochemical assays. Our data are consistent with the concept that AF produces its effects through a multitarget mechanism that mainly affects the redox metabolism and the mitochondrial functions and results into severe ER stress. Results are discussed in the context of the current mechanistic knowledge existing on AF. Redox proteomics allows to underline cell adaptation mechanisms in response to Auranofin treatment in ovarian cancer cells. BRCA1 is one of the major candidates of the ovarian cancer cell adaptation to Auranofin treatment. Auranofin alters the oxidative phosphorylation and mitochondrial protein import machinery. TRAP1 C501 modulates Auranofin toxicity. Auranofin induces severe stress of the endoplasmic reticulum.
Collapse
Affiliation(s)
- Giovanni Chiappetta
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France.
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134, Florence, Italy.
| | - Fiorella Faienza
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Xhesika Limaj
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France
| | - Salvatore Rizza
- Redox Signaling and Oxidative Stress Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Luigi Messori
- Metmed Lab, Department of Chemistry, University of Florence, via della lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy; Redox Signaling and Oxidative Stress Group, Danish Cancer Society Research Center, Copenhagen, Denmark; Center for Healthy Aging, University of Copenhagen, Denmark
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Joelle Vinh
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France
| |
Collapse
|
123
|
Bondeson DP, Paolella BR, Asfaw A, Rothberg MV, Skipper TA, Langan C, Mesa G, Gonzalez A, Surface LE, Ito K, Kazachkova M, Colgan WN, Warren A, Dempster JM, Krill-Burger JM, Ericsson M, Tang AA, Fung I, Chambers ES, Abdusamad M, Dumont N, Doench JG, Piccioni F, Root DE, Boehm J, Hahn WC, Mannstadt M, McFarland JM, Vazquez F, Golub TR. Phosphate dysregulation via the XPR1-KIDINS220 protein complex is a therapeutic vulnerability in ovarian cancer. NATURE CANCER 2022; 3:681-695. [PMID: 35437317 PMCID: PMC9246846 DOI: 10.1038/s43018-022-00360-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/04/2022] [Indexed: 12/13/2022]
Abstract
Despite advances in precision medicine, the clinical prospects for patients with ovarian and uterine cancers have not substantially improved. Here, we analyzed genome-scale CRISPR/Cas9 loss-of-function screens across 851 human cancer cell lines and found that frequent overexpression of SLC34A2 – encoding a phosphate importer – is correlated to sensitivity to loss of the phosphate exporter XPR1 in vitro and in vivo. In patient-derived tumor samples, we observed frequent PAX8-dependent overexpression of SLC34A2, XPR1 copy number amplifications, and XPR1 mRNA overexpression. Mechanistically, in SLC34A2-high cancer cell lines, genetic or pharmacologic inhibition of XPR1-dependent phosphate efflux leads to the toxic accumulation of intracellular phosphate. Finally, we show that XPR1 requires the novel partner protein KIDINS220 for proper cellular localization and activity, and that disruption of this protein complex results in acidic vacuolar structures preceding cell death. These data point to the XPR1:KIDINS220 complex and phosphate dysregulation as a therapeutic vulnerability in ovarian cancer. Golub and colleagues identify the phosphate exporter XPR1 as a therapeutic vulnerability in ovarian and uterine cancers, and show that phosphate efflux inhibition reduces tumor cell viability through accumulation of intracellular phosphate.
Collapse
Affiliation(s)
| | - Brenton R Paolella
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Merck Research Laboratories, Cambridge, MA, USA
| | - Adhana Asfaw
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Carly Langan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gabriel Mesa
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Lauren E Surface
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Kentaro Ito
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | | | | | - Andrew A Tang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Iris Fung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Mai Abdusamad
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nancy Dumont
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Federica Piccioni
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Merck Research Laboratories, Cambridge, MA, USA
| | - David E Root
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesse Boehm
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Harvard Medical School, Boston, MA, USA.,Departments of Pediatric and Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Michael Mannstadt
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | | | - Todd R Golub
- Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Harvard Medical School, Boston, MA, USA. .,Departments of Pediatric and Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
124
|
CT-Based Radiomics and Deep Learning for BRCA Mutation and Progression-Free Survival Prediction in Ovarian Cancer Using a Multicentric Dataset. Cancers (Basel) 2022; 14:cancers14112739. [PMID: 35681720 PMCID: PMC9179845 DOI: 10.3390/cancers14112739] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/15/2022] [Accepted: 05/29/2022] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Build predictive radiomic models for early relapse and BRCA mutation based on a multicentric database of high-grade serous ovarian cancer (HGSOC) and validate them in a test set coming from different institutions. METHODS Preoperative CTs of patients with HGSOC treated at four referral centers were retrospectively acquired and manually segmented. Hand-crafted features and deep radiomics features were extracted respectively by dedicated software (MODDICOM) and a dedicated convolutional neural network (CNN). Features were selected with and without prior harmonization (ComBat harmonization), and models were built using different machine learning algorithms, including clinical variables. RESULTS We included 218 patients. Radiomic models showed low performance in predicting both BRCA mutation (AUC in test set between 0.46 and 0.59) and 1-year relapse (AUC in test set between 0.46 and 0.56); deep learning models demonstrated similar results (AUC in the test of 0.48 for BRCA and 0.50 for relapse). The inclusion of clinical variables improved the performance of the radiomic models to predict BRCA mutation (AUC in the test set of 0.74). CONCLUSIONS In our multicentric dataset, representative of a real-life clinical scenario, we could not find a good radiomic predicting model for PFS and BRCA mutational status, with both traditional radiomics and deep learning, but the combination of clinical and radiomic models improved model performance for the prediction of BRCA mutation. These findings highlight the need for standardization through the whole radiomic pipelines and robust multicentric external validations of results.
Collapse
|
125
|
Sriramkumar S, Sood R, Huntington TD, Ghobashi AH, Vuong TT, Metcalfe TX, Wang W, Nephew KP, O'Hagan HM. Platinum-induced mitochondrial OXPHOS contributes to cancer stem cell enrichment in ovarian cancer. J Transl Med 2022; 20:246. [PMID: 35641987 PMCID: PMC9153190 DOI: 10.1186/s12967-022-03447-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/18/2022] [Indexed: 01/06/2023] Open
Abstract
Background Platinum based agents—cisplatin and carboplatin in combination with taxanes are used for the treatment of ovarian cancer (OC) patients. However, the majority of OC patients develop recurrent, platinum resistant disease that is uniformly fatal. Platinum treatment enriches for chemoresistant aldehyde dehydrogenase (ALDH) + ovarian cancer stem cells (OCSCs), which contribute to tumor recurrence and disease relapse. Acquired platinum resistance also includes metabolic reprograming and switching to oxidative phosphorylation (OXPHOS). Chemosensitive cells rely on glycolysis while chemoresistant cells have the ability to switch between glycolysis and OXPHOS, depending on which pathway drives a selective advantage for growth and chemoresistance. High expression of genes involved in OXPHOS and high production of mitochondrial ROS are characteristics of OCSCs, suggesting that OCSCs favor OXPHOS over glycolysis. Based on connections between OCSCs, chemoresistance and OXPHOS, we hypothesize that platinum treatment induces changes in metabolism that contribute to platinum-induced enrichment of OCSCs. Methods The effect of cisplatin on mitochondrial activity was assessed by JC1 staining and expression of OXPHOS genes by RT-qPCR. Cisplatin-induced changes in Sirtuin 1 (SIRT1) levels and activity were assessed by western blot. Small molecule inhibitors of mitochondrial complex I and SIRT1 were used to determine if their enzymatic activity contributes to the platinum-induced enrichment of OCSCs. The percentage of ALDH + OCSCs in OC cells and tumor tissue from xenograft models across different treatment conditions was analyzed using ALDEFLUOR assay and flow cytometry. Results We demonstrate that platinum treatment increases mitochondrial activity. Combined treatment of platinum agents and OXPHOS inhibitors blocks the platinum-induced enrichment of ALDH + OCSCs in vitro and in vivo. Furthermore, platinum treatment increases SIRT1 levels and subsequent deacetylase activity, which likely contributes to the increase in platinum-induced mitochondrial activity. Conclusions These findings on metabolic pathways altered by platinum-based chemotherapy have uncovered key targets that can be exploited therapeutically to block the platinum-induced enrichment of OCSCs, ultimately improving the survival of OC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03447-y.
Collapse
Affiliation(s)
- Shruthi Sriramkumar
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Riddhi Sood
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Genome, Cell and Developmental Biology, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA
| | - Thomas D Huntington
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Ahmed H Ghobashi
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Genome, Cell and Developmental Biology, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA
| | - Truc T Vuong
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Tara X Metcalfe
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Weini Wang
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Kenneth P Nephew
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.,Genome, Cell and Developmental Biology, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA.,Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology and Physiology, Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heather M O'Hagan
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA. .,Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA. .,Genome, Cell and Developmental Biology, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA. .,Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA. .,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
126
|
Pavlič R, Gjorgoska M, Rižner TL. Model Cell Lines and Tissues of Different HGSOC Subtypes Differ in Local Estrogen Biosynthesis. Cancers (Basel) 2022; 14:cancers14112583. [PMID: 35681563 PMCID: PMC9179372 DOI: 10.3390/cancers14112583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Ovarian cancer (OC) comprises a heterogeneous group of hormone-dependent diseases with very high mortality. Estrogens have been shown to promote the progression of OC; however, their exact role in OC subtypes remains unknown. Here, we investigated the local estrogen biosynthesis in OC. We performed targeted transcriptomics and estrogen metabolism analyses in high-grade serous OC (HGSOC) cell lines that differed in chemoresistance status and compared these data with publicly available transcriptome and proteome data for HGSOC tissues. In HGSOC cells, estrogen metabolism decreased with increasing chemoresistance. In highly chemoresistant cells and platinum-resistant HGSOC tissues, HSD17B14 expression was increased. Proteome data showed differential levels of HSD17B10, SULT1E1, CYP1B1, and NQO1 between the four HGSOC subtypes. Our results confirm that estrogen biosynthesis differs between different HGSOC cell models and possibly between different HGSOC subtypes. Such differentially expressed enzymes have potential as targets in the search of new treatment options. Abstract Ovarian cancer (OC) is highly lethal and heterogeneous. Several hormones are involved in OC etiology including estrogens; however, their role in OC is not completely understood. Here, we performed targeted transcriptomics and estrogen metabolism analyses in high-grade serous OC (HGSOC), OVSAHO, Kuramochi, COV632, and immortalized normal ovarian epithelial HIO-80 cells. We compared these data with public transcriptome and proteome data for the HGSOC tissues. In all model systems, high steroid sulfatase expression and weak/undetected aromatase (CYP19A1) expression indicated the formation of estrogens from the precursor estrone-sulfate (E1-S). In OC cells, the metabolism of E1-S to estradiol was the highest in OVSAHO, followed by Kuramochi and COV362 cells, and decreased with increasing chemoresistance. In addition, higher HSD17B14 and CYP1A2 expressions were observed in highly chemoresistant COV362 cells and platinum-resistant tissues compared to those in HIO-80 cells and platinum-sensitive tissues. The HGSOC cell models differed in HSD17B10, CYP1B1, and NQO1 expression. Proteomic data also showed different levels of HSD17B10, CYP1B1, NQO1, and SULT1E1 between the four HGSOC subtypes. These results suggest that different HGSOC subtypes form different levels of estrogens and their metabolites and that the estrogen-biosynthesis-associated targets should be further studied for the development of personalized treatment.
Collapse
|
127
|
Zhao LQ, Sun W, Zhang P, Gao W, Fang CY, Zheng AW. MFAP2 aggravates tumor progression through activating FOXM1/β-catenin-mediated glycolysis in ovarian cancer. Kaohsiung J Med Sci 2022; 38:772-780. [PMID: 35546486 DOI: 10.1002/kjm2.12546] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/21/2022] [Accepted: 03/18/2022] [Indexed: 11/08/2022] Open
Abstract
Ovarian cancer is one of the most common gynecological tumors that seriously endanger the health and quality of life of women. Microfibril-associated protein 2 (MFAP2) has been demonstrated to play crucial roles in the development of multiple tumors. However, the function of MFAP2 in ovarian cancer remains unclear. In this study, we found that MFAP2 was upregulated in ovarian cancer and cells and was positively correlated with FOXM1 and glycolysis-related genes. The results of Cell Count Kit-8, colony formation, and flow cytometry assays indicated that MFAP2 promoted cell proliferation. In addition, MFAP2 promotes cell proliferation, glucose uptake, lactate production; increases ATP levels, extracellular acidification ratio, and oxygen consumption ratio in ovarian cancer cells and increases the expression of glycolytic proteins. Further mechanistic analysis suggests that MFAP2 promotes FOXM1/β-catenin-mediated glycolysis signaling in ovarian cancer cells. Knockdown of MFAP2 inhibits ovarian cancer xenograft tumor growth and expression of Ki-67, MFAP2, FOXM1, GLUT1, HK2, and β-catenin in mice. In conclusion, MFAP2 promotes cell proliferation and glycolysis by modulating the FOXM1/β-catenin signaling pathway in ovarian cancer, which may offer a fresh insight into the treatment of ovarian cancer in the glycolysis pathway.
Collapse
Affiliation(s)
- Ling-Qin Zhao
- Department of Gynecologic Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wei Sun
- Department of Gynecologic Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Ping Zhang
- Department of Gynecologic Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wen Gao
- Department of Gynecologic Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Chen-Yan Fang
- Department of Gynecologic Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Ai-Wen Zheng
- Department of Gynecologic Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
128
|
The Multiple Pharmacologic Functions and Mechanisms of Action of Guizhi Fuling Formulation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6813421. [PMID: 35529925 PMCID: PMC9076289 DOI: 10.1155/2022/6813421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/09/2022] [Indexed: 11/17/2022]
Abstract
Objectives Guizhi Fuling Formulation (GZFL), a traditional Chinese medical formulation, consists of Cinnamomi Ramulus, Paeoniae Radix Alba (or Paeoniae Radix Rubra), Moutan Cortex, Persicae Semen, and Poria, with multiple therapeutic functions such as sedation, antitumor activity, anti-inflammation, and neuroprotection. However, its clinical applications remain relatively fragmented, and the underlying mechanisms of GZFL in different diseases are still not very certain. Further research and summary in both application and mechanisms remain to be needed for human health and the best use of GZFL. Therefore, we summarized the multiple pharmacologic effects and possible mechanisms of action of GZFL according to recent 17 years of research. Methods We retrieved four English and two Chinese databases using these keywords (the formulation name or its synonyms) and searched articles written in English from January 2006 up to February 2022. Key Findings. GZFL exhibits multiple pharmacologic advantages in gynecologic diseases and other expanding diseases such as cancer, blood, and vascular disease, renal failure, inflammation, and brain injury. Possibly due to its diverse bioactive components and pharmacologic activities, GZFL could target the multiple signaling pathways involved in regulating blood circulation, inflammatory and immune factors, proliferation, apoptosis, and so on. Conclusion This review suggests that GZFL displays promising therapeutic effects for many kinds of diseases, which have been beyond the scope of the original prescription for gynecologic diseases. In this way, we wish to provide a reference and recommendation for further preclinic and clinic studies.
Collapse
|
129
|
A Redoxable Mn Porphyrin, MnTnBuOE-2-PyP5+, Synergizes with Carboplatin in Treatment of Chemoresistant Ovarian Cell Line. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9664636. [PMID: 35898616 PMCID: PMC9313984 DOI: 10.1155/2022/9664636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/29/2022] [Indexed: 12/20/2022]
Abstract
We have employed a redox-active MnP (MnTnBuOE-2-PyP5+, Mn(III) meso-tetrakis (N-n-butoxyethylpyridinium-2-yl) porphyrin) frequently identified as superoxide dismutase mimic or BMX-001, to explore the redox status of normal ovarian cell in relation to two ovarian cancer cell lines: OV90 human serous ovarian cancer cell and chemotherapy-resistant OV90 cell (OVCD). We identified that OVCD cells are under oxidative stress due to high hydrogen peroxide (H2O2) levels and low glutathione peroxidase and thioredoxin 1. Furthermore, OVCD cells have increased glycolysis activity and mitochondrial respiration when compared to immortalized ovarian cells (hTER7) and parental cancer cells (OV90). Our goal was to study how ovarian cell growth depends upon the redox state of the cell; hence, we used MnP (BMX-001), a redox-active MnSOD mimetic, as a molecular tool to alter ovarian cancer redox state. Interestingly, OVCD cells preferentially uptake MnP relative to OV90 cells which led to increased inhibition of cell growth, glycolytic activity, OXPHOS, and ATP, in OVCD cells. These effects were further increased when MnP was combined with carboplatin. The effects were discussed with regard to the elevation in H2O2 levels, increased oxidative stress, and reduced Nrf2 levels and its downstream targets when cells were exposed to either MnP or MnP/carboplatin. It is significant to emphasize that MnP protects normal ovarian cell line, hTER7, against carboplatin toxicity. Our data demonstrate that the addition of MnP-based redox-active drugs may be used (via increasing excessively the oxidative stress of serous ovarian cancer cells) to improve cancer patients' chemotherapy outcomes, which develop resistance to platinum-based drugs.
Collapse
|
130
|
He WP, Yang GP, Yang ZX, Shen HW, You ZS, Yang GF. Maelstrom promotes tumor metastasis through regulation of FGFR4 and epithelial-mesenchymal transition in epithelial ovarian cancer. J Ovarian Res 2022; 15:55. [PMID: 35513870 PMCID: PMC9074322 DOI: 10.1186/s13048-022-00992-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Increasing evidence has indicated that Maelstrom (MAEL) plays an oncogenic role in various human carcinomas. However, the exact function and mechanisms by which MAEL acts in epithelial ovarian cancer (EOC) remain unclear. RESULTS This study demonstrated that MAEL was frequently overexpressed in EOC tissues and cell lines. Overexpression of MAEL was positively correlated with the histological grade of tumors, FIGO stage, and pT/pN/pM status (p < 0.05), and it also acted as an independent predictor of poor patient survival (p < 0.001). Ectopic overexpression of MAEL substantially promoted invasiveness/metastasis and induced epithelial-mesenchymal transition (EMT), whereas silencing MAEL by short hairpin RNA effectively inhibited its oncogenic function and attenuated EMT. Further study demonstrated that fibroblast growth factor receptor 4 (FGFR4) was a critical downstream target of MAEL in EOC, and the expression levels of FGFR4 were significantly associated with MAEL. (P < 0.05). CONCLUSION Our findings suggest that overexpression of MAEL plays a crucial oncogenic role in the development and progression of EOC through the upregulation of FGFR4 and subsequent induction of EMT, and also provide new insights on its potential as a therapeutic target for EOC.
Collapse
Affiliation(s)
- Wei-Peng He
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Gui-Ping Yang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Zun-Xian Yang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Hong-Wei Shen
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Ze-Shan You
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Guo-Fen Yang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China.
| |
Collapse
|
131
|
Understanding the Experience of Canadian Women Living with Ovarian Cancer through the Every Woman Study TM. Curr Oncol 2022; 29:3318-3340. [PMID: 35621661 PMCID: PMC9139742 DOI: 10.3390/curroncol29050271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
The Every Woman StudyTM: Canadian Edition is the most comprehensive study to date exploring patient-reported experiences of ovarian cancer (OC) on a national scale. An online survey conducted in Fall 2020 included individuals diagnosed with OC in Canada, reporting responses from 557 women from 11 Canadian provinces/territories. Median age at diagnosis was 54 (11−80), 61% were diagnosed between 2016−2020, 59% were stage III/IV and all subtypes of OC were represented. Overall, 23% had a family history of OC, 75% had genetic testing and 19% reported having a BRCA1/2 mutation. Most (87%) had symptoms prior to diagnosis. A timely diagnosis of OC (≤3 months from first presentation with symptoms) was predicted by age (>50) or abdominal pain/persistent bloating as the primary symptom. Predictors of an acute diagnosis (<1 month) included region, ER/urgent care doctor as first healthcare provider or stage III/IV disease. Regional differences in genetic testing, treatments and clinical trial participation were also noted. Respondents cited substantial physical, emotional, practical and financial impacts of an OC diagnosis. Our national survey has revealed differences in the pathway to diagnosis and post-diagnostic care among Canadian women with OC, with region, initial healthcare provider, specific symptoms and age playing key roles. We have identified many opportunities to improve both clinical and supportive care of OC patients across the country.
Collapse
|
132
|
Chehade H, Tedja R, Ramos H, Bawa TS, Adzibolosu N, Gogoi R, Mor G, Alvero AB. Regulatory Role of the Adipose Microenvironment on Ovarian Cancer Progression. Cancers (Basel) 2022; 14:cancers14092267. [PMID: 35565396 PMCID: PMC9101128 DOI: 10.3390/cancers14092267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Adipocytes or fat cells are integral part of the ovarian tumor microenvironment. Secreted factors from adipocytes, as well as direct cell-to-cell interaction with ovarian cancer cells have been shown to directly support ovarian tumor progression. Elucidating the molecular pathways involved is crucial in the identification of relevant targets. Abstract The tumor microenvironment of ovarian cancer is the peritoneal cavity wherein adipose tissue is a major component. The role of the adipose tissue in support of ovarian cancer progression has been elucidated in several studies from the past decades. The adipocytes, in particular, are a major source of factors, which regulate all facets of ovarian cancer progression such as acquisition of chemoresistance, enhanced metastatic potential, and metabolic reprogramming. In this review, we summarize the relevant studies, which highlight the role of adipocytes in ovarian cancer progression and offer insights into unanswered questions and possible future directions of research.
Collapse
Affiliation(s)
- Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA; (H.C.); (R.T.); (H.R.); (T.S.B.); (N.A.); (R.G.); (G.M.)
| | - Roslyn Tedja
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA; (H.C.); (R.T.); (H.R.); (T.S.B.); (N.A.); (R.G.); (G.M.)
- Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Harry Ramos
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA; (H.C.); (R.T.); (H.R.); (T.S.B.); (N.A.); (R.G.); (G.M.)
| | - Tejeshwar Singh Bawa
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA; (H.C.); (R.T.); (H.R.); (T.S.B.); (N.A.); (R.G.); (G.M.)
| | - Nicholas Adzibolosu
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA; (H.C.); (R.T.); (H.R.); (T.S.B.); (N.A.); (R.G.); (G.M.)
| | - Radhika Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA; (H.C.); (R.T.); (H.R.); (T.S.B.); (N.A.); (R.G.); (G.M.)
- Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA; (H.C.); (R.T.); (H.R.); (T.S.B.); (N.A.); (R.G.); (G.M.)
- Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Ayesha B. Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA; (H.C.); (R.T.); (H.R.); (T.S.B.); (N.A.); (R.G.); (G.M.)
- Karmanos Cancer Institute, Detroit, MI 48201, USA
- Correspondence:
| |
Collapse
|
133
|
Shi L, Sun HJ, Zeng JJ, Liang ZQ, Lin YH, Huang SN, Zeng JH, Yang L, Chen H, Luo J, Wei KL. Evaluation of miR-141-3p over-expression in ovarian cancer. ELECTRON J BIOTECHN 2022. [DOI: 10.1016/j.ejbt.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
134
|
Wei YF, Sun ML, Wen ZY, Liu FH, Liu YS, Yan S, Qin X, Gao S, Li XQ, Zhao YH, Gong TT, Wu QJ. Pre-diagnosis meat intake and cooking method and ovarian cancer survival: results from the Ovarian Cancer Follow-Up Study (OOPS). Food Funct 2022; 13:4653-4663. [PMID: 35373791 DOI: 10.1039/d1fo03825g] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Objectives: The relationships between pre-diagnosis meat intake and ovarian cancer (OC) survival were limited and controversial. To date, no study has taken account of cooking methods. Thus, we aimed to firstly clarify these associations based on the Ovarian Cancer Follow-Up Study. Methods: This prospective cohort study, including 853 OC patients between 2015 and 2020, was conducted to examine the aforementioned associations. All women completed a food frequency questionnaire. Deaths were ascertained up to March 31, 2021 via medical records and active follow-up. We used the Cox proportional hazards model to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). Results: During the median follow-up of 37.17 months, 130 women died. Pre-diagnosis fish and seafood intake was associated with better survival (HRT3 vs. T1 = 0.46, 95% CI = 0.26-0.82, p trend <0.05), whereas processed red meat (HR = 1.54, 95% CI = 1.04-2.26) and a high frequency of fried fish intake (HR = 1.49, 95% CI = 1.03-2.16) were associated with worse survival than consuming none. After considering the interaction of cooking methods, we found that compared with the lowest tertile of fish and seafood intake and almost no fried fish cooking, women with the highest tertile of intake and almost no fried fish cooking had better survival (HR = 0.35, 95% CI = 0.13-0.92). Additionally, compared with the lowest tertile of fish and seafood intake and almost no baked fish cooking, women with the lowest tertile of intake and consuming baked fish had worse survival (HR = 3.75, 95% CI = 1.53-9.15). Conclusions: Pre-diagnosis fish and seafood intake was associated with better OC survival, whereas processed red meat intake was associated with worse survival. Cooking methods, especially for fried or baked fish, may play interaction effects with fish intake on OC survival.
Collapse
Affiliation(s)
- Yi-Fan Wei
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming-Li Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Zhao-Yan Wen
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang-Hua Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ya-Shu Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shi Yan
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xue Qin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Song Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Xiu-Qin Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yu-Hong Zhao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
135
|
Choi HY, Ahn JH, Kwon H, Yim JH, Lee D, Choi JH. Citromycin Isolated from the Antarctic Marine-Derived Fungi, Sporothrix sp., Inhibits Ovarian Cancer Cell Invasion via Suppression of ERK Signaling. Mar Drugs 2022; 20:md20050275. [PMID: 35621926 PMCID: PMC9143255 DOI: 10.3390/md20050275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
Recently, microorganisms and their metabolites in the Antarctic marine environment have attracted attention as useful sources for novel therapeutics, including anticancer drugs. Here, we investigated the effects of citromycin, isolated from the Antarctic marine-derived fungus, Sporothrix sp., on human ovarian cancer cells. Citromycin inhibited the migration and invasion of human ovarian cancer SKOV3 and A2780 cells, but had no cytotoxic activity against them. Additionally, it inhibited the expression of epithelial–mesenchymal transition (EMT) markers and the activation of matrix metalloproteinase (MMP)-2 and MMP9. Moreover, extracellular signal-regulated kinase (ERK)-1/2 signaling was inhibited after citromycin treatment, and the ectopic expression of ERK negated the anti-invasive activity of citromycin. Our findings suggest that citromycin inhibits the migration and invasion of human ovarian cancer cells by downregulating the expression levels of EMT markers and MMP-2/9 via inhibition of the ERK1/2 pathway.
Collapse
Affiliation(s)
- He Yun Choi
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Korea;
| | - Ji-Hye Ahn
- Department of Oriental Pharmacy, Woosuk University, Jeonju 55338, Korea;
| | - Haeun Kwon
- Department of Plant Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (H.K.); (D.L.)
| | - Joung Han Yim
- Korea Polar Research Institute, Korea Ocean Research and Development Institute, Incheon 21990, Korea;
| | - Dongho Lee
- Department of Plant Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (H.K.); (D.L.)
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Korea;
- College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
- Correspondence:
| |
Collapse
|
136
|
TLR4 Agonist and Hypoxia Synergistically Promote the Formation of TLR4/NF-κB/HIF-1α Loop in Human Epithelial Ovarian Cancer. Anal Cell Pathol (Amst) 2022; 2022:4201262. [PMID: 35464826 PMCID: PMC9023210 DOI: 10.1155/2022/4201262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 02/11/2022] [Accepted: 03/15/2022] [Indexed: 12/04/2022] Open
Abstract
Inflammation and hypoxia are involved in numerous cancer progressions. Reportedly, the toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) pathway and hypoxia-inducible factor-1α (HIF-1α) are activated and closely related to the chemoresistance and poor prognosis of epithelial ovarian cancer (EOC). However, the potential correlation between TLR4/NF-κB and HIF-1α remains largely unknown in EOC. In our study, the possible positive correlation among TLR4, NF-κB, and HIF-1α proteins was investigated in the EOC tissues. Our in vitro results demonstrated that LPS can induce and activate HIF-1α through the TLR4/NF-κB signaling in A2780 and SKOV3 cells. Moreover, hypoxia-induced TLR4 expression and the downstream transcriptional activity of NF-κB were HIF-1α-dependent. The cross talk between the TLR4/NF-κB signaling pathway and HIF-1α was also confirmed in the nude mice xenograft model. Therefore, we first proposed the formation of a TLR4/NF-κB/HIF-1α loop in EOC. The positive feedback loop enhanced the susceptibility and responsiveness to inflammation and hypoxia, which synergistically promote the initiation and progression of EOC. The novel mechanism may act as a future therapeutic candidate for the treatment of EOC.
Collapse
|
137
|
Alexandrova E, Salvati A, Pecoraro G, Lamberti J, Melone V, Sellitto A, Rizzo F, Giurato G, Tarallo R, Nassa G, Weisz A. Histone Methyltransferase DOT1L as a Promising Epigenetic Target for Treatment of Solid Tumors. Front Genet 2022; 13:864612. [PMID: 35495127 PMCID: PMC9043692 DOI: 10.3389/fgene.2022.864612] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
The histone lysine methyltransferase DOT1L (DOT1-like histone lysine methyltransferase) is responsible for the epigenetic regulation of gene expression through specific methylation of lysine79 residue of histone H3 (H3K79) in actively transcribed genes. Its normal activity is crucial for embryonic development and adult tissues functions, whereas its aberrant functioning is known to contribute to leukemogenesis. DOT1L is the only lysine methyltransferase that does not contain a SET domain, which is a feature that allowed the development of selective DOT1L inhibitors that are currently investigated in Phase I clinical trials for cancer treatment. Recently, abnormal expression of this enzyme has been associated with poor survival and increased aggressiveness of several solid tumors. In this review evidences of aberrant DOT1L expression and activity in breast, ovarian, prostate, colon, and other solid tumors, and its relationships with biological and clinical behavior of the disease and response to therapies, are summarized. Current knowledge of the structural basis of DOT1L ability to regulate cell proliferation, invasion, plasticity and stemness, cell cycle progression, cell-to-cell signaling, epithelial-to-mesenchymal transition, and chemoresistance, through cooperation with several molecular partners including noncoding RNAs, is also reviewed. Finally, available options for the treatment of therapeutically challenging solid tumors by targeting DOT1L are discussed.
Collapse
Affiliation(s)
- Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Medical Genomics Program and Division of Oncology, AOU “S. Giovanni di Dio e Ruggi d’Aragona”, University of Salerno, Salerno, Italy
| | - Giovanni Pecoraro
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Jessica Lamberti
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Viola Melone
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Assunta Sellitto
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
- *Correspondence: Giovanni Nassa, ; Alessandro Weisz,
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Medical Genomics Program and Division of Oncology, AOU “S. Giovanni di Dio e Ruggi d’Aragona”, University of Salerno, Salerno, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
- *Correspondence: Giovanni Nassa, ; Alessandro Weisz,
| |
Collapse
|
138
|
Electroporation and Electrochemotherapy in Gynecological and Breast Cancer Treatment. Molecules 2022; 27:molecules27082476. [PMID: 35458673 PMCID: PMC9026735 DOI: 10.3390/molecules27082476] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/14/2022] [Accepted: 04/10/2022] [Indexed: 12/24/2022] Open
Abstract
Gynecological carcinomas affect an increasing number of women and are associated with poor prognosis. The gold standard treatment plan is mainly based on surgical resection and subsequent chemotherapy with cisplatin, 5-fluorouracil, anthracyclines, or taxanes. Unfortunately, this treatment is becoming less effective and is associated with many side effects that negatively affect patients’ physical and mental well-being. Electroporation based on tumor exposure to electric pulses enables reduction in cytotoxic drugs dose while increasing their effectiveness. EP-based treatment methods have received more and more interest in recent years and are the subject of a large number of scientific studies. Some of them show promising therapeutic potential without using any cytotoxic drugs or molecules already present in the human body (e.g., calcium electroporation). This literature review aims to present the fundamental mechanisms responsible for the course of EP-based therapies and the current state of knowledge in the field of their application in the treatment of gynecological neoplasms.
Collapse
|
139
|
Li Y, Zhang Y, Yang Q, Zhou X, Guo Y, Ding F, Liu Z, Luo A. Silencing of FANCI Promotes DNA Damage and Sensitizes Ovarian Cancer Cells to Carboplatin. Curr Cancer Drug Targets 2022; 22:591-602. [PMID: 35362384 DOI: 10.2174/1568009622666220331091709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/31/2022] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ovarian cancer (OVCA) has unique epigenetic alterations and defects in homologous recombination (HR). Despite initial sensitivity to platinum-based chemotherapy, HR dysfunctional tumors eventually acquire drug resistance. Fanconi anemia (FA) is characterized by bone marrow failure (BMF) and a reduced ability to eradicate DNA interstrand cross-links (ICL). However, the mechanism of chemoresistance mediated by FANCI was unclear in OVCA. OBJECTIVE We explore to identify whether FANCI was involved in chemoresistance in OVCA. METHODS FANCI expression and epigenetic alterations were analyzed, respectively, using TIMER and cBioPortal. The correlation between FANCI expression and the survival of OVCA patients was analyzed using Kaplan-Meier Plotter, GSE63885 and TCGA-OVCA database. FANCI expression in OVCA was detected by immunohistochemistry. Cell proliferation, migration, and invasion in FANCI inhibiting cells were assessed by CCK8 and Transwell. Apoptosis and DNA damage were examined by flow cytometry and immunofluorescence. Meanwhile, the activity of caspase 3/7 was detected by Caspase-Glo® 3/7 kit. In addition, the expression of FANCI, γH2AX, and apoptosis effectors was examined by western blot. RESULTS FANCI has copy number variations (CNVs) in OVCA. The high expression of FANCI in OVCA patients was associated with poor survival. Moreover, FANCI expression was correlated with the response to chemotherapy in OVCA. FANCI expression in OVCA cells was induced by carboplatin in a time-dependent manner. Silencing of FANCI had no effect on cell proliferation, but it hindered OVCA cell migration and invasion. Mechanically, knockdown of FANCI enhanced DNA damage induced apoptosis through CHK1/2-P53-P21 pathway. CONCLUSION FANCI may be a potential therapeutic target for OVCA patients.
Collapse
Affiliation(s)
- Yuqing Li
- State Key Lab of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Yanan Zhang
- State Key Lab of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Qi Yang
- State Key Lab of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Xuantong Zhou
- State Key Lab of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Yuanyuan Guo
- State Key Lab of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Fang Ding
- State Key Lab of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Zhihua Liu
- State Key Lab of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Aiping Luo
- State Key Lab of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| |
Collapse
|
140
|
Li H, Luo F, Jiang X, Zhang W, Xiang T, Pan Q, Cai L, Zhao J, Weng D, Li Y, Dai Y, Sun F, Yang C, Huang Y, Yang J, Tang Y, Han Y, He M, Zhang Y, Song L, Xia JC. CircITGB6 promotes ovarian cancer cisplatin resistance by resetting tumor-associated macrophage polarization toward the M2 phenotype. J Immunother Cancer 2022; 10:jitc-2021-004029. [PMID: 35277458 PMCID: PMC8919471 DOI: 10.1136/jitc-2021-004029] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2022] [Indexed: 12/28/2022] Open
Abstract
Background Platinum resistance is a major challenge in the clinical treatment of advanced ovarian cancer (OC). Accumulating evidence shows that the tumor-promotive M2 macrophage is linked to the limiting chemotherapy efficacy of multiple malignancies including OC. Circular RNAs (circRNAs) are a novel class of non-coding RNAs which function as the critical regulator in biological process of cancer. However, their impact on macrophage polarization and chemoresistance of OC remain unclear. Methods Platinum-resistant circRNAs were screened using circRNA deep sequencing and validated using in situ hybridization in OC tissues with or without platinum resistance. The role of circITGB6 in inducing cisplatin (CDDP) resistance was evaluated by clone formation, immunofluorescence and annexin V assays in vitro, and by intraperitoneal tumor model in vivo. The mechanism underlying circITGB6-mediated tumor-associated macrophage (TAM) polarization into M2 phenotype was investigated using RNA pull-down, luciferase reporter, electrophoretic mobility shift, RNA binding protein immunoprecipitation (RIP), ELISA and immunofluorescence assays. Results We identified that a novel circRNA, circITGB6, robustly elevated in tumor tissues and serums from patients with OC with platinum resistance, was correlated with poor prognosis. circITGB6 overexpression promoted an M2 macrophage-dependent CDDP resistance in both vivo and vitro. Mechanistic research determined that circITGB6 directly interacted with IGF2BP2 and FGF9 mRNA to form a circITGB6/IGF2BP2/FGF9 RNA–protein ternary complex in the cytoplasm, thereby stabilizing FGF9 mRNA and inducing polarization of TAMs toward M2 phenotype. Importantly, blocking M2 macrophage polarization with an antisense oligonucleotide targeting circITGB6 markedly reversed the circITGB6-induced CDDP resistance of OC in vivo. Conclusions This study reveals a novel mechanism for platinum resistance in OC and demonstrates that circITGB6 may serve as a potential prognostic marker and a therapeutic target for patients with OC.
Collapse
Affiliation(s)
- Han Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Fan Luo
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xingyu Jiang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Weijing Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tong Xiang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qiuzhong Pan
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Liming Cai
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Zhao
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Desheng Weng
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yue Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuhu Dai
- Department of Orthopaedic Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Fengze Sun
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chaopin Yang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yue Huang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jieying Yang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yan Tang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yulong Han
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Mian He
- Department of Gynecology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Yanna Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Libing Song
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jian-Chuan Xia
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
141
|
Anderson KG, Oda SK, Bates BM, Burnett MG, Rodgers Suarez M, Ruskin SL, Greenberg PD. Engineering adoptive T cell therapy to co-opt Fas ligand-mediated death signaling in ovarian cancer enhances therapeutic efficacy. J Immunother Cancer 2022; 10:jitc-2021-003959. [PMID: 35264436 PMCID: PMC8915280 DOI: 10.1136/jitc-2021-003959] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2022] [Indexed: 01/23/2023] Open
Abstract
Background In the USA, more than 50% of patients with ovarian cancer die within 5 years of diagnosis, highlighting the need for therapeutic innovations. Mesothelin (MSLN) is a candidate immunotherapy target; it is overexpressed by ovarian tumors and contributes to malignant/invasive phenotypes, making tumor antigen loss disadvantageous. We previously showed that MSLN-specific T cell receptor (TCR)-engineered T cells preferentially accumulate within established tumors, delay tumor growth, and significantly prolong survival in the ID8VEGF mouse model that replicates many aspects of human disease. However, T cell persistence and antitumor activity were not sustained. We therefore focused on Fas/FasL signaling that can induce activation-induced cell death, an apoptotic mechanism that regulates T cell expansion. Upregulation of FasL by tumor cells and tumor vasculature has been detected in the tumor microenvironment (TME) of human and murine ovarian cancers, can induce apoptosis in infiltrating, Fas (CD95) receptor-expressing lymphocytes, and can protect ovarian cancers from tumor-infiltrating lymphocytes. Methods To overcome potential FasL-mediated immune evasion and enhance T cell responses, we generated an immunomodulatory fusion protein (IFP) containing the Fas extracellular binding domain fused to a 4-1BB co-stimulatory domain, rather than the natural death domain. Murine T cells were engineered to express an MSLN-specific TCR (TCR1045), alone or with the IFP, transferred into ID8VEGF tumor-bearing mice and evaluated for persistence, proliferation, cytokine production and efficacy. Human T cells were similarly engineered to express an MSLN-specific TCR (TCR530) alone or with a truncated Fas receptor or a Fas-4-1BB IFP and evaluated for cytokine production and tumor lysis. Results Relative to murine T cells expressing only TCR1045, T cells expressing both TCR1045 and a Fas-4-1BB IFP preferentially persisted in the TME of tumor-bearing mice, with improved T cell proliferation and survival. Moreover, TCR1045/IFP+ T cells significantly prolonged survival in tumor-bearing mice, compared with TCR1045-only T cells. Human T cells expressing TCR530 and a Fas-4-1BB IFP exhibit enhanced functional activity and viability compared with cells with only TCR530. Conclusions As many ovarian tumors overexpress FasL, an IFP that converts the Fas-mediated death signal into pro-survival and proliferative signals may be used to enhance engineered adoptive T cell therapy for patients.
Collapse
Affiliation(s)
- Kristin G Anderson
- Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Shannon K Oda
- Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Breanna M Bates
- Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Madison G Burnett
- Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Susan L Ruskin
- Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Philip D Greenberg
- Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA .,Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
142
|
Wang Z, Chen W, Zuo L, Xu M, Wu Y, Huang J, Zhang X, Li Y, Wang J, Chen J, Wang H, Sun H. The Fibrillin-1/VEGFR2/STAT2 signaling axis promotes chemoresistance via modulating glycolysis and angiogenesis in ovarian cancer organoids and cells. Cancer Commun (Lond) 2022; 42:245-265. [PMID: 35234370 PMCID: PMC8923131 DOI: 10.1002/cac2.12274] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/31/2021] [Accepted: 02/07/2022] [Indexed: 12/14/2022] Open
Abstract
Background Chemotherapy resistance is a primary reason of ovarian cancer therapy failure; hence it is important to investigate the underlying mechanisms of chemotherapy resistance and develop novel potential therapeutic targets. Methods RNA sequencing of cisplatin‐resistant and ‐sensitive (chemoresistant and chemosensitive, respectively) ovarian cancer organoids was performed, followed by detection of the expression level of fibrillin‐1 (FBN1) in organoids and clinical specimens of ovarian cancer. Subsequently, glucose metabolism, angiogenesis, and chemosensitivity were analyzed in structural glycoprotein FBN1‐knockout cisplatin‐resistant ovarian cancer organoids and cell lines. To gain insights into the specific functions and mechanisms of action of FBN1 in ovarian cancer, immunoprecipitation, silver nitrate staining, mass spectrometry, immunofluorescence, Western blotting, and Fӧrster resonance energy transfer‐fluorescence lifetime imaging analyses were performed, followed by in vivo assays using vertebrate model systems of nude mice and zebrafish. Results FBN1 expression was significantly enhanced in cisplatin‐resistant ovarian cancer organoids and tissues, indicating that FBN1 might be a key factor in chemoresistance of ovarian cancer. We also discovered that FBN1 sustained the energy stress and induced angiogenesis in vitro and in vivo, which promoted the cisplatin‐resistance of ovarian cancer. Knockout of FBN1 combined with treatment of the antiangiogenic drug apatinib improved the cisplatin‐sensitivity of ovarian cancer cells. Mechanistically, FBN1 mediated the phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2) at the Tyr1054 residue, which activated its downstream focal adhesion kinase (FAK)/protein kinase B (PKB or AKT) pathway, induced the phosphorylation of signal transducer and activator of transcription 2 (STAT2) at the tyrosine residue 690 (Tyr690), promoted the nuclear translocation of STAT2, and ultimately altered the expression of genes associated with STAT2‐mediated angiogenesis and glycolysis. Conclusions The FBN1/VEGFR2/STAT2 signaling axis may induce chemoresistance of ovarian cancer cells by participating in the process of glycolysis and angiogenesis. The present study suggested a novel FBN1‐targeted therapy and/or combination of FBN1 inhibition and antiangiogenic drug for treating ovarian cancer.
Collapse
Affiliation(s)
- Ziliang Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, P. R. China.,Institute of Cancer Research and Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Wei Chen
- Department of Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Ling Zuo
- Institute of Cancer Research and Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Midie Xu
- Department of Pathology and Biobank, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Yong Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Jiami Huang
- Institute of Cancer Research and Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Xu Zhang
- Institute of Cancer Research and Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Yongheng Li
- Institute of Cancer Research and Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Jing Wang
- Department of Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Jing Chen
- Institute of Cancer Research and Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Husheng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, P. R. China
| | - Huizhen Sun
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, P. R. China
| |
Collapse
|
143
|
Single cell-derived spheroids capture the self-renewing subpopulations of metastatic ovarian cancer. Cell Death Differ 2022; 29:614-626. [PMID: 34845371 PMCID: PMC8901794 DOI: 10.1038/s41418-021-00878-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 11/24/2022] Open
Abstract
High Grade Serous Ovarian cancer (HGSOC) is a major unmet need in oncology, due to its precocious dissemination and the lack of meaningful human models for the investigation of disease pathogenesis in a patient-specific manner. To overcome this roadblock, we present a new method to isolate and grow single cells directly from patients' metastatic ascites, establishing the conditions for propagating them as 3D cultures that we refer to as single cell-derived metastatic ovarian cancer spheroids (sMOCS). By single cell RNA sequencing (scRNAseq) we define the cellular composition of metastatic ascites and trace its propagation in 2D and 3D culture paradigms, finding that sMOCS retain and amplify key subpopulations from the original patients' samples and recapitulate features of the original metastasis that do not emerge from classical 2D culture, including retention of individual patients' specificities. By enabling the enrichment of uniquely informative cell subpopulations from HGSOC metastasis and the clonal interrogation of their diversity at the functional and molecular level, this method provides a powerful instrument for precision oncology in ovarian cancer.
Collapse
|
144
|
Shim JI, Ryu JY, Jeong SY, Cho YJ, Choi JJ, Hwang JR, Choi JY, Sa JK, Lee JW. Combination effect of poly (ADP-ribose) polymerase inhibitor and DNA demethylating agents for treatment of epithelial ovarian cancer. Gynecol Oncol 2022; 165:270-280. [DOI: 10.1016/j.ygyno.2022.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 11/04/2022]
|
145
|
USP13 promotes development and metastasis of high-grade serous ovarian carcinoma in a novel mouse model. Oncogene 2022; 41:1974-1985. [PMID: 35173307 PMCID: PMC8956511 DOI: 10.1038/s41388-022-02224-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 01/07/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
Abstract
Epithelial ovarian cancer is the most lethal gynecologic malignancy and one of the most common causes of cancer mortality among women worldwide. Ubiquitin-Specific Peptidase 13 (USP13) gene copy is strongly amplified in human epithelial ovarian cancer, and high USP13 expression is correlated with poor survival outcomes. Yet, its pathological contribution to ovarian tumorigenesis remains unknown. We crossed a conditional Usp13 overexpressing knock-in mouse with a conditional knockout of Trp53 and Pten mouse and generated a novel ovarian cancer genetically engineered mouse model (GEMM), which closely recapitulates the genetic changes driving ovarian cancer in humans. Overexpression of USP13 with deletion of Trp53 and Pten in murine ovarian surface epithelium accelerated ovarian tumorigenesis and led to decreased survival in mice. Notably, USP13 greatly enhanced peritoneal metastasis of ovarian tumors with frequent development of hemorrhagic ascites. The primary and metastatic tumors exhibited morphology and clinical behavior similar to human high-grade serous ovarian cancer. Co-inhibition of USP13 and AKT significantly decreased the viability of the primary murine ovarian cancer cells isolated from the GEMM. USP13 also increased the tumorigenic and metastatic abilities of primary murine ovarian cancer cells in a syngeneic mouse study. These findings suggest a critical role of USP13 in ovarian cancer development and reveal USP13 as a potential therapeutic target for ovarian cancer.
Collapse
|
146
|
Oncogenic Events Dictate the Types and Locations of Gynecological Malignancies Originating from Krt8+ Mesothelial and Müllerian-Derived Epithelial Cells. Cancers (Basel) 2022; 14:cancers14030841. [PMID: 35159108 PMCID: PMC8834519 DOI: 10.3390/cancers14030841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Ovarian and uterine cancers are the most common gynecological malignancies in women. The early detection, prevention, and treatment of these gynecological cancers can benefit from a better understanding of how tumor-initiating cells in them are formed from their corresponding target cell populations in the female reproductive system. To study this, we utilized a genetic approach in mice to introduce driver mutations commonly found in these cancers to Keratin 8 positive (K8+) mesothelial and epithelial cells in the ovary, fallopian tube, and uterus. We found that p53-loss appears to preferentially affect K8+ epithelial cells, leading to the development of uterine and ovarian malignancies, whereas PTEN-loss may preferentially affect mesothelial cells, leading to the development of ovarian endometrioid malignancies or adenoma on the fallopian tube surface. Collectively, our data suggest that oncogenic driver mutations may dominantly determine the locations and types of gynecological malignancies developed from K8+ mesothelial and epithelial cells in the female reproductive system. Abstract Ovarian and uterine cancers are the most prevalent types of gynecological malignancies originating from mesothelial and/or Müllerian-derived epithelial cells. Recent genomic studies have identified common mutations in them that affect signaling pathways such as p53, PTEN/PI3K, RAS, and WNT pathways. However, how these mutations and their corresponding deregulated pathways affect gynecological cancer development from their cells-of-origin remains largely elusive. To address this, we performed the intrabursal injection of Cre-expressing adenovirus under the control of Krt8 promoter (Ad-K8-Cre) to mice carrying combinations of various conditional alleles for cancer genes. We found that Ad-K8-Cre specifically targeted mesothelial cells, including ovarian surface epithelial (OSE) cells (mainly the LGR5+ subset of OSE cells) and mesothelial cells lining the fallopian tube (FT) serosa; the injected Ad-K8-Cre also targeted Müllerian-derived epithelial cells, including FT epithelial cells and uterine endometrial epithelial cells. The loss of p53 may preferentially affect Müllerian-derived epithelial cells, leading to the development of uterine and ovarian malignancies, whereas PTEN-loss may preferentially affect mesothelial cells, leading to the development of ovarian endometrioid malignancies (upon KRAS-activation or APC-loss) or adenoma on the FT surface (upon DICER-loss). Overall, our data suggest that different Krt8+ mesothelial and epithelial cell types in the female reproductive system may have different sensitivities toward oncogenic mutations and, as a result, oncogenic events may dominantly determine the locations and types of the gynecological malignancies developed from them.
Collapse
|
147
|
Hong J, Xie Z, Yang Z, Yang F, Liao H, Rao S, Huang X. Inactivation of Wnt-LRP5 signaling suppresses the proliferation and migration of ovarian cancer cells. Transl Cancer Res 2022; 10:2277-2285. [PMID: 35116545 PMCID: PMC8797788 DOI: 10.21037/tcr-20-3462] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/05/2021] [Indexed: 11/15/2022]
Abstract
Background Ovarian cancer (OCa) is the most lethal gynecological malignant tumor, with few or no specific symptoms in its early stage. There are many signaling pathways involved in the process of OCa progression, among which the highly complex Wnt signaling pathway plays a unique role in the occurrence and development of OCa because of its functions of regulating gene expression, cell proliferation, migration, and invasion. Lipoprotein associated receptor protein 5/6 (LRP5/6) binds to activate this key pathway. Therefore, it is very important to study the mechanism of Wnt-LRP5 signaling pathway in the proliferation and migration of OCa. Methods In the present study, we have investigated the role of Wnt-LRP5 signaling pathway in OCa proliferation and migration for the first time using the dominant negative plasmid of LRP5 (DN-LRP5) and human OCa cells HO8910PM plus in a mouse model. Results Our data showed inactivation of LRP5 resulted in shift of epithelial-mesenchymal transition (EMT), rearrangement of the cytoskeleton, lowered activity of pro-proliferation and pro-migration cancer signaling pathways including Akt, p38 and NF-κB, eventually decreased proliferation and migration of OCa cells HO8910PM in vitro. Moreover, in vivo OCa-DN-LRP5 mouse model developed significantly smaller tumors as determined by inoculation of HO8910PM-DN-LRP5 cells into nude mice. Conclusions Collectively, our results demonstrate the dominant role of Wnt-LRP5 in OCa proliferation and migration and its potential as a valuable therapeutic target.
Collapse
Affiliation(s)
- Jing Hong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Zeyu Xie
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Zhihua Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Fangyao Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Hai Liao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Shuquan Rao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Xinhe Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
148
|
Gershenson DM, Miller A, Brady WE, Paul J, Carty K, Rodgers W, Millan D, Coleman RL, Moore KN, Banerjee S, Connolly K, Secord AA, O'Malley DM, Dorigo O, Gaillard S, Gabra H, Slomovitz B, Hanjani P, Farley J, Churchman M, Ewing A, Hollis RL, Herrington CS, Huang HQ, Wenzel L, Gourley C. Trametinib versus standard of care in patients with recurrent low-grade serous ovarian cancer (GOG 281/LOGS): an international, randomised, open-label, multicentre, phase 2/3 trial. Lancet 2022; 399:541-553. [PMID: 35123694 PMCID: PMC8819271 DOI: 10.1016/s0140-6736(21)02175-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/12/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Low-grade serous carcinoma of the ovary or peritoneum is characterised by MAPK pathway aberrations and its reduced sensitivity to chemotherapy relative to high-grade serous carcinoma. We compared the MEK inhibitor trametinib to physician's choice standard of care in patients with recurrent low-grade serous carcinoma. METHODS This international, randomised, open-label, multicentre, phase 2/3 trial was done at 84 hospitals in the USA and UK. Eligible patients were aged 18 years or older with recurrent low-grade serous carcinoma and measurable disease, as defined by Response Evaluation Criteria In Solid Tumors version 1.1, had received at least one platinum-based regimen, but not all five standard-of-care drugs, and had received an unlimited number of previous regimens. Patients with serous borderline tumours or tumours containing low-grade serous and high-grade serous carcinoma were excluded. Eligible patients were randomly assigned (1:1) to receive either oral trametinib 2 mg once daily (trametinib group) or one of five standard-of-care treatment options (standard-of-care group): intravenous paclitaxel 80 mg/m2 by body surface area on days 1, 8, and 15 of every 28-day cycle; intravenous pegylated liposomal doxorubicin 40-50 mg/m2 by body surface area once every 4 weeks; intravenous topotecan 4 mg/m2 by body surface area on days 1, 8, and 15 of every 28-day cycle; oral letrozole 2·5 mg once daily; or oral tamoxifen 20 mg twice daily. Randomisation was stratified by geographical region (USA or UK), number of previous regimens (1, 2, or ≥3), performance status (0 or 1), and planned standard-of-care regimen. The primary endpoint was investigator-assessed progression-free survival while receiving randomised therapy, as assessed by imaging at baseline, once every 8 weeks for 15 months, and then once every 3 months thereafter, in the intention-to-treat population. Safety was assessed in patients who received at least one dose of study therapy. This trial is registered with ClinicalTrials.gov, NCT02101788, and is active but not recruiting. FINDINGS Between Feb 27, 2014, and April 10, 2018, 260 patients were enrolled and randomly assigned to the trametinib group (n=130) or the standard-of-care group (n=130). At the primary analysis, there were 217 progression-free survival events (101 [78%] in the trametinib group and 116 [89%] in the standard-of-care group). Median progression-free survival in the trametinib group was 13·0 months (95% CI 9·9-15·0) compared with 7·2 months (5·6-9·9) in the standard-of-care group (hazard ratio 0·48 [95% CI 0·36-0·64]; p<0·0001). The most frequent grade 3 or 4 adverse events in the trametinib group were skin rash (17 [13%] of 128), anaemia (16 [13%]), hypertension (15 [12%]), diarrhoea (13 [10%]), nausea (12 [9%]), and fatigue (ten [8%]). The most frequent grade 3 or 4 adverse events in the standard-of-care group were abdominal pain (22 [17%]), nausea (14 [11%]), anaemia (12 [10%]), and vomiting (ten [8%]). There were no treatment-related deaths. INTERPRETATION Trametinib represents a new standard-of-care option for patients with recurrent low-grade serous carcinoma. FUNDING NRG Oncology, Cancer Research UK, Target Ovarian Cancer, and Novartis.
Collapse
Affiliation(s)
- David M Gershenson
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Austin Miller
- NRG Oncology, Clinical Trial Development Division, Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - William E Brady
- NRG Oncology, Clinical Trial Development Division, Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - James Paul
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Karen Carty
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - William Rodgers
- New York Presbyterian/Queens, Department of Pathology, Weill Medical College of Cornell University, Flushing, NY, USA
| | - David Millan
- Queen Elizabeth University Hospital, Glasgow, UK
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kathleen N Moore
- Stephenson Cancer Center at the University of Oklahoma, Oklahoma City, OK, USA
| | - Susana Banerjee
- The Royal Marsden Hospital NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | | | | | - David M O'Malley
- The Ohio State University and the James Cancer Center, Columbus, OH, USA
| | - Oliver Dorigo
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | | | - Hani Gabra
- Surgery and Cancer, Imperial College London, London, UK
| | - Brian Slomovitz
- Division ofGynecologic Oncology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | | | - John Farley
- St Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Michael Churchman
- Nicola Murray Centre for Ovarian Cancer Research, Institute of Genetics and Cancer, Western General Hospital, Edinburgh, UK
| | - Ailith Ewing
- MRC Human Genetics Unit and CRUK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Robert L Hollis
- Nicola Murray Centre for Ovarian Cancer Research, Institute of Genetics and Cancer, Western General Hospital, Edinburgh, UK
| | - C Simon Herrington
- Nicola Murray Centre for Ovarian Cancer Research, Institute of Genetics and Cancer, Western General Hospital, Edinburgh, UK
| | - Helen Q Huang
- NRG Oncology, Clinical Trial Development Division, Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Lari Wenzel
- Medicine and Public Health, University of California at Irvine, Irvine, CA, USA
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Institute of Genetics and Cancer, Western General Hospital, Edinburgh, UK
| |
Collapse
|
149
|
Belotti Y, Lim EH, Lim CT. The Role of the Extracellular Matrix and Tumor-Infiltrating Immune Cells in the Prognostication of High-Grade Serous Ovarian Cancer. Cancers (Basel) 2022; 14:404. [PMID: 35053566 PMCID: PMC8773831 DOI: 10.3390/cancers14020404] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer is the eighth global leading cause of cancer-related death among women. The most common form is the high-grade serous ovarian carcinoma (HGSOC). No further improvements in the 5-year overall survival have been seen over the last 40 years since the adoption of platinum- and taxane-based chemotherapy. Hence, a better understanding of the mechanisms governing this aggressive phenotype would help identify better therapeutic strategies. Recent research linked onset, progression, and response to treatment with dysregulated components of the tumor microenvironment (TME) in many types of cancer. In this study, using bioinformatic approaches, we identified a 19-gene TME-related HGSOC prognostic genetic panel (19 prognostic genes (PLXNB2, HMCN2, NDNF, NTN1, TGFBI, CHAD, CLEC5A, PLXNA1, CST9, LOXL4, MMP17, PI3, PRSS1, SERPINA10, TLL1, CBLN2, IL26, NRG4, and WNT9A) by assessing the RNA sequencing data of 342 tumors available in the TCGA database. Using machine learning, we found that specific patterns of infiltrating immune cells characterized each risk group. Furthermore, we demonstrated the predictive potential of our risk score across different platforms and its improved prognostic performance compared with other gene panels.
Collapse
Affiliation(s)
- Yuri Belotti
- Institute for Health Innovation and Technology, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore;
| | - Elaine Hsuen Lim
- Division of Medical Oncology, National Cancer Center Singapore, 11 Hospital Drive, Singapore 169610, Singapore;
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore;
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
150
|
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: 0.5] [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
|