1
|
Perrone C, Angioli R, Luvero D, Giannini A, Di Donato V, Cuccu I, Muzii L, Raspagliesi F, Bogani G. Targeting BRAF pathway in low-grade serous ovarian cancer. J Gynecol Oncol 2024; 35:e104. [PMID: 38768941 PMCID: PMC11262891 DOI: 10.3802/jgo.2024.35.e104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/14/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Mutations in genes encoding for proteins along the RAS-RAF-MEK-ERK pathway have been detected in a variety of tumor entities including ovarian carcinomas. In the recent years, several inhibitors of this pathway have been developed, whose antitumor potential is currently being assessed in different clinical trials. Low grade serous ovarian carcinoma, is a rare gynecological tumor which shows favorable overall survival, compared to the general ovarian cancer population, but worrying resistance to conventional chemotherapies. The clinical behavior of low grade serous ovarian carcinoma reflects the different gene profile compared to high-grade serous carcinoma: KRAS/BRAF mutations. BRAF inhibitors as single agents were approved for the treatment of BRAF mutated tumors. Nevertheless, many patients face progressive disease. The understanding of the mechanisms of resistance to BRAF inhibitors therapy and preclinical studies showing that BRAF and mitogen-activated protein kinase kinase (MEK) inhibitors combined therapy delays the onset of resistance compared to BRAF inhibitor single agent, led to the clinical investigation of combined therapy. The aim of this paper is to review the efficacy and safety of the combination of BRAF plus MEK inhibitors on ovarian carcinomas, in particularly focusing on low grade serous ovarian carcinoma.
Collapse
Affiliation(s)
- Chiara Perrone
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Roberto Angioli
- Department of Gynecology, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Daniela Luvero
- Department of Gynecology, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Andrea Giannini
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Violante Di Donato
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ilaria Cuccu
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ludovico Muzii
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Raspagliesi
- Gynecologic Oncologic Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Giorgio Bogani
- Gynecologic Oncologic Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| |
Collapse
|
2
|
Murawski M, Jagodziński A, Bielawska-Pohl A, Klimczak A. Complexity of the Genetic Background of Oncogenesis in Ovarian Cancer-Genetic Instability and Clinical Implications. Cells 2024; 13:345. [PMID: 38391958 PMCID: PMC10886918 DOI: 10.3390/cells13040345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Ovarian cancer is a leading cause of death among women with gynecological cancers, and is often diagnosed at advanced stages, leading to poor outcomes. This review explores genetic aspects of high-grade serous, endometrioid, and clear-cell ovarian carcinomas, emphasizing personalized treatment approaches. Specific mutations such as TP53 in high-grade serous and BRAF/KRAS in low-grade serous carcinomas highlight the need for tailored therapies. Varying mutation prevalence across subtypes, including BRCA1/2, PTEN, PIK3CA, CTNNB1, and c-myc amplification, offers potential therapeutic targets. This review underscores TP53's pivotal role and advocates p53 immunohistochemical staining for mutational analysis. BRCA1/2 mutations' significance as genetic risk factors and their relevance in PARP inhibitor therapy are discussed, emphasizing the importance of genetic testing. This review also addresses the paradoxical better prognosis linked to KRAS and BRAF mutations in ovarian cancer. ARID1A, PIK3CA, and PTEN alterations in platinum resistance contribute to the genetic landscape. Therapeutic strategies, like restoring WT p53 function and exploring PI3K/AKT/mTOR inhibitors, are considered. The evolving understanding of genetic factors in ovarian carcinomas supports tailored therapeutic approaches based on individual tumor genetic profiles. Ongoing research shows promise for advancing personalized treatments and refining genetic testing in neoplastic diseases, including ovarian cancer. Clinical genetic screening tests can identify women at increased risk, guiding predictive cancer risk-reducing surgery.
Collapse
Affiliation(s)
- Marek Murawski
- 1st Clinical Department of Gynecology and Obstetrics, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Adam Jagodziński
- 1st Clinical Department of Gynecology and Obstetrics, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Aleksandra Bielawska-Pohl
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.B.-P.); (A.K.)
| | - Aleksandra Klimczak
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.B.-P.); (A.K.)
| |
Collapse
|
3
|
Abstract
The RAS family of proteins is among the most frequently mutated genes in human malignancies. In ovarian cancer (OC), the most lethal gynecological malignancy, RAS, especially KRAS mutational status at codons 12, 13, and 61, ranges from 6-65% spanning different histo-types. Normally RAS regulates several signaling pathways involved in a myriad of cellular signaling cascades mediating numerous cellular processes like cell proliferation, differentiation, invasion, and death. Aberrant activation of RAS leads to uncontrolled induction of several downstream signaling pathways such as RAF-1/MAPK (mitogen-activated protein kinase), PI3K phosphoinositide-3 kinase (PI3K)/AKT, RalGEFs, Rac/Rho, BRAF (v-Raf murine sarcoma viral oncogene homolog B), MEK1 (mitogen-activated protein kinase kinase 1), ERK (extracellular signal-regulated kinase), PKB (protein kinase B) and PKC (protein kinase C) involved in cell proliferation as well as maintenance pathways thereby driving tumorigenesis and cancer cell propagation. KRAS mutation is also known to be a biomarker for poor outcome and chemoresistance in OC. As a malignancy with several histotypes showing varying histopathological characteristics, we focus on reviewing recent literature showcasing the involvement of oncogenic RAS in mediating carcinogenesis and chemoresistance in OC and its subtypes.
Collapse
Affiliation(s)
- Lubna Therachiyil
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
| | - Anjana Anand
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
| | | | | | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
| | - Shahab Uddin
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
| |
Collapse
|
4
|
Sohn EJ. PIK3R3, a regulatory subunit of PI3K, modulates ovarian cancer stem cells and ovarian cancer development and progression by integrative analysis. BMC Cancer 2022; 22:708. [PMID: 35761259 PMCID: PMC9238166 DOI: 10.1186/s12885-022-09807-7] [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: 03/23/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background Ovarian cancer is the most lethal gynecologic disease and is one of the most commonly diagnosed cancers among women worldwide. The phosphatidylinositol 3-kinase (PI3K) family plays an important regulatory role in various cancer signaling pathways, including those involved in ovarian cancer development; however, its exact function remains to be fully understood. We conducted this study to understand the role of P13K in the molecular mechanisms underlying ovarian cancer development. Methods To determine the differential gene expression of phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3), a regulatory subunit of PI3K, in normal, tumor, and metastatic ovary tissues, TNM plotter analysis was performed. The microarray dataset GSE53759 was downloaded from Gene Expression Omnibus. ROC plotter analysis was conducted to understand the potential of PIK3R3 as a predictive marker for effectiveness of therapy in ovarian cancer. muTarget was used to identify mutations that alter PIK3R3 expression in ovarian cancer. To determine the interacting partners for PIK3R3 in ovarian tissues, the interactome-atlas tool was used. The Kyoto encyclopedia of genes and genomes (KEGG) analysis was conducted to identify the pathways in which these interacting partners were primarily enriched. Results PIK3R3 was overexpressed in ovarian and metastatic tumors. Elevated PIK3R3 levels were observed in ovarian cancer stem cells, wherein inhibiting PIK3R3 expression significantly reduced the size of ovarian cancer spheroids. Treatment of ovarian cancer stem cells with PF-04691502 (10 μM), an inhibitor of both PI3K and mTOR kinases, also reduced the size of spheroids and the level of OCT4. PIK3R3 was highly expressed in ovarian cancer with several somatic mutations and was predicted better outcomes in patients undergoing Avastin® chemotherapy using bioinformatic tool. Protein interaction analysis showed that PIK3R3 interacts with 157 genes, including GRB2, EGFR, ERBB3, PTK2, HCK, IGF1R, YES1, and PIK3CA, in the ovary. KEGG enrichment analysis revealed that the interacting partners of PIK3R3 are involved in the ErbB signaling pathway, proteoglycans in cancer, FoxO, prolactin, chemokine, and insulin signaling pathways. Conclusions PIK3R3 plays a pivotal role in ovarian cancer development and is therefore a potential candidate for developing novel therapeutic approaches. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09807-7.
Collapse
Affiliation(s)
- Eun Jung Sohn
- Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
5
|
Study on the Function and Mechanism of miR-585-3p Inhibiting the Progression of Ovarian Cancer Cells by Targeting FSCN1 to Block the MAPK Signaling Pathway. Anal Cell Pathol (Amst) 2022; 2022:1732365. [PMID: 35602576 PMCID: PMC9122712 DOI: 10.1155/2022/1732365] [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: 11/24/2021] [Revised: 03/22/2022] [Accepted: 04/09/2022] [Indexed: 11/17/2022] Open
Abstract
Ovarian cancer (OC) is the leading cause of death for women diagnosed with gynecological cancer. Studies have shown that dysregulated miRNA expression is related to various cancers, including OC. Here, we aimed to explore the biological function and mechanism of miR-585-3p in the occurrence and development of OC. The expression level of miR-585-3p was found to be low in OC tissues and cells. We analyzed the biological function of miR-585-3p in OC through in vitro cell experiments. The results indicated that overexpression of miR-585-3p inhibited the proliferation, invasion, and migration of SW626 cells, while low expression of miR-585-3p had the opposite effect in SKOV3 cells. We then screened the target genes of miR-585-3p through miRDB database and detected the expression of target genes in OC cells. FSCN1 was found to be most significantly upregulated in OC cells. Dual-luciferase reporter assays revealed FSCN1 as a potential target of miR-585-3p. Western blot analysis showed that miR-585-3p targeted FSCN1 to inhibit protein phosphorylation of ERK. In vivo animal experiments also confirmed that miR-585-3p targets FSCN1 to inhibit tumor growth and block the MAPK signaling pathway. In summary, miR-585-3p inhibits the proliferation, migration, and invasion of OC cells by targeting FSCN1, and its mechanism of action may be achieved by inhibiting the activation of the MAPK signaling pathway. miR-585-3p may serve as a potential biomarker and therapeutic target for OC.
Collapse
|
6
|
Zhu JW, Charkhchi P, Akbari MR. Potential clinical utility of liquid biopsies in ovarian cancer. Mol Cancer 2022; 21:114. [PMID: 35545786 PMCID: PMC9092780 DOI: 10.1186/s12943-022-01588-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the most lethal gynecologic malignancy worldwide. One of the main challenges in the management of OC is the late clinical presentation of disease that results in poor survival. Conventional tissue biopsy methods and serological biomarkers such as CA-125 have limited clinical applications. Liquid biopsy is a novel sampling method that analyzes distinctive tumour components released into the peripheral circulation, including circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), cell-free RNA (cfRNA), tumour-educated platelets (TEPs) and exosomes. Increasing evidence suggests that liquid biopsy could enhance the clinical management of OC by improving early diagnosis, predicting prognosis, detecting recurrence, and monitoring response to treatment. Capturing the unique tumour genetic landscape can also guide treatment decisions and the selection of appropriate targeted therapies. Key advantages of liquid biopsy include its non-invasive nature and feasibility, which allow for serial sampling and longitudinal monitoring of dynamic tumour changes over time. In this review, we outline the evidence for the clinical utility of each liquid biopsy component and review the advantages and current limitations of applying liquid biopsy in managing ovarian cancer. We also highlight future directions considering the current challenges and explore areas where more studies are warranted to elucidate its emerging clinical potential.
Collapse
Affiliation(s)
- Jie Wei Zhu
- Women's College Research Institute, Women's College Hospital, University of Toronto, 76 Grenville St, Toronto, ON, M5S 1B2, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Parsa Charkhchi
- Women's College Research Institute, Women's College Hospital, University of Toronto, 76 Grenville St, Toronto, ON, M5S 1B2, Canada
| | - Mohammad R Akbari
- Women's College Research Institute, Women's College Hospital, University of Toronto, 76 Grenville St, Toronto, ON, M5S 1B2, Canada.
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
7
|
Bonsang-Kitzis H, Panchbhaya N, Bats AS, Pujade-Lauraine E, Pautier P, Ngô C, Le Frère-Belda MA, Kalbacher E, Floquet A, Berton-Rigaud D, Lefeuvre-Plesse C, Fabbro M, Ray-Coquard I, Lécuru F. Surgical Implications of Advanced Low-Grade Serous Ovarian Cancer: Analysis of the Database of the Tumeurs Malignes Rares Gynécologiques Network. Cancers (Basel) 2022; 14:cancers14092345. [PMID: 35565475 PMCID: PMC9105599 DOI: 10.3390/cancers14092345] [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: 03/19/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 11/19/2022] Open
Abstract
Simple Summary Low-grade serous carcinoma is a recent entity. The surgical management of advanced stages is modeled on that of high-grade tumors, with the use of neoadjuvant chemotherapy in the case of carcinosis not amenable to complete primary resection. We retrospectively analyzed data from the French national network dedicated to rare gynecologic tumors. We compared disease extension, surgical characteristics, postoperative course and survival after primary surgery vs. interval debulking. Carcinosis was more extended in the case of neoadjuvant chemotherapy. However, chemotherapy did not reduce surgical complexity, nor late postoperative morbidity. Surprisingly, progression-free and overall survival were similar after complete macroscopic or minimal resection (residuals < 2.5 mm). Survival was similar in the case of residuals ≥2.5 mm or more and nonoperated patients. Neoadjuvant chemotherapy does not improve the resectability of advanced low-grade serous cancers. Primary cytoreduction with complete or with minimal residuals should be preferred when feasible. Abstract The surgical specificities of advanced low-grade serous ovarian carcinoma (LGSOC) have been little investigated. Our objective was to describe surgical procedures/complications in primary (PDS) compared to interval debulking surgery (neoadjuvant chemotherapy and interval debulking surgery, NACT-IDS) and to assess the survival (progression-free (PFS) and overall survival (OS)) in patients with advanced LGSOC. We retrospectively analyzed advanced LGSOC from a nationwide registry (January 2000 to July 2017). A total of 127 patients were included (48% PDS and 35% NACT-IDS). Peritoneal carcinomatosis was more severe (p = 0.01 to 0.0001, according to sites), surgery more complex (p = 0.03) and late postoperative morbidity more frequent (p = 0.03) and more severe in the NACT-IDS group. PFS and OS were similar in patients with CC0 and CC1 residual disease after PDS or IDS. Prognosis was poorest for NACT-IDS patients with CC2/CC3 resection (PFS: HR = 2.31, IC95% (1.3–4.58); p = 0.005; OS: HR = 4.98, IC95% (1.59–15.61); p = 0.006). NACT has no benefit in terms of surgical outputs in patients with advanced LGSOC. Patients with complete resection or minimal residual disease (CC0 and CC1) have similar prognoses. On the other hand, patients with CC2 and more residual disease have similar survival rates compared to nonoperated patients. Primary cytoreduction with complete or with minimal residuals should be preferred when feasible.
Collapse
Affiliation(s)
- Hélène Bonsang-Kitzis
- Gynecological and Breast Surgery and Cancerology Center, RAMSAY-Générale de Santé, Hôpital Privé des Peupliers, 75013 Paris, France; (H.B.-K.); (C.N.)
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
| | - Nabilah Panchbhaya
- Department of Obsterics and Gynecology, Hôpital Lariboisière, Assistance Publique—Hôpitaux de Paris, 75020 Paris, France;
| | - Anne-Sophie Bats
- Department of Breast and Gynecological Surgical Oncology, Hôpital Européen Georges Pompidou, Assistance Publique—Hôpitaux de Paris, 75015 Paris, France;
- School of Medicine, Université de Paris, 75005 Paris, France
| | - Eric Pujade-Lauraine
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- School of Medicine, Université de Paris, 75005 Paris, France
- Women Cancer Center and Clinical Research, Hôpital Hôtel-Dieu, Assistance Publique—Hôpitaux de Paris, 75006 Paris, France
| | - Patricia Pautier
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Department of Medical Oncology, Institut Gustave Roussy, 94000 Villejuif, France
| | - Charlotte Ngô
- Gynecological and Breast Surgery and Cancerology Center, RAMSAY-Générale de Santé, Hôpital Privé des Peupliers, 75013 Paris, France; (H.B.-K.); (C.N.)
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
| | - Marie-Aude Le Frère-Belda
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Department of Pathology, Hôpital Européen Georges Pompidou, Assistance Publique—Hôpitaux de Paris, 75015 Paris, France
| | - Elsa Kalbacher
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Department of Medical Oncology, Hôpital Jean Minjoz, 25000 Besançon, France
| | - Anne Floquet
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Department of Medical Oncology, Institut Bergonié, 33000 Bordeaux, France
| | - Dominique Berton-Rigaud
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Department of Medical Oncology, Institut de Cancérologie de l’Ouest-René Gauducheau, 44000 Saint Herblain, France
| | - Claudia Lefeuvre-Plesse
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Department of Medical Oncology, Centre Eugène Marquis, 35000 Rennes, France
| | - Michel Fabbro
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Department of Medical Oncology, Institut Régional du Cancer de Montpellier, 33000 Montpellier, France
| | - Isabelle Ray-Coquard
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Department of Medical Oncology, Centre Léon Bérard, 69000 Lyon, France
- School of Medicine, Claude Bernard University, 69000 Lyon, France
| | - Fabrice Lécuru
- GINECO/TMRG Network, 75008 Paris, France; (E.P.-L.); (P.P.); (M.-A.L.F.-B.); (E.K.); (A.F.); (D.B.-R.); (C.L.-P.); (M.F.); (I.R.-C.)
- Women Cancer Center and Clinical Research, Hôpital Hôtel-Dieu, Assistance Publique—Hôpitaux de Paris, 75006 Paris, France
- Department of Breast, Gynecology and Reconstructive Surgery, Institut Curie, 75005 Paris, France
- Correspondence:
| |
Collapse
|
8
|
Bartoletti M, Musacchio L, Giannone G, Tuninetti V, Bergamini A, Scambia G, Lorusso D, Valabrega G, Mangili G, Puglisi F, Pignata S. Emerging molecular alterations leading to histology-specific targeted therapies in ovarian cancer beyond PARP inhibitors. Cancer Treat Rev 2021; 101:102298. [PMID: 34634660 DOI: 10.1016/j.ctrv.2021.102298] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 01/12/2023]
Abstract
After more than 30 years of a one-size-fits-all approach in the management of advanced ovarian cancer, in 2018 the SOLO1 trial results have introduced a new era of personalized medicine. A deeper knowledge of ovarian cancer biology and the development of new drugs targeting specific molecular pathways have led to biomarker-driven phase 3 trials with practice changing results. Thereafter, platinum-based combinations are no longer the only therapeutic options available in first line setting and poly-ADP ribose polymerase inhibitors maintenance therapy has become the mainstay in patients with tumor harboring a homologous recombination defect. However, most of the recent therapeutic breakthroughs regard high grade serous carcinoma, the most frequent ovarian cancer subtype, and only few improvements have occurred in the management of less common histotypes. Moving towards the next challenges, we aimed to investigate and review new potential molecular targets in ovarian cancer, according to histotype, starting from promising molecular drivers and matched drugs that have been investigated in early and late-stage clinical trials or conceptualized in preclinical studies.
Collapse
Affiliation(s)
- M Bartoletti
- Department of Medicine (DAME), University of Udine, Udine, Italy; Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano (PN), Italy
| | - L Musacchio
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - G Giannone
- Candiolo Cancer Institute, FPO- IRCCS, Candiolo (TO), Italy; Department of Oncology, University of Turin, Torino, Piemonte, Italy
| | - V Tuninetti
- Candiolo Cancer Institute, FPO- IRCCS, Candiolo (TO), Italy; Department of Oncology, University of Turin, Torino, Piemonte, Italy
| | - A Bergamini
- Department of Obstetrics and Gynecology, IRCCS, San Raffaele Hospital, Milan, Italy
| | - G Scambia
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Life Science and Public Health, Catholic University of Sacred Heart Largo Agostino Gemelli, Rome, Italy
| | - D Lorusso
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Life Science and Public Health, Catholic University of Sacred Heart Largo Agostino Gemelli, Rome, Italy
| | - G Valabrega
- Candiolo Cancer Institute, FPO- IRCCS, Candiolo (TO), Italy; Department of Oncology, University of Turin, Torino, Piemonte, Italy
| | - G Mangili
- Department of Obstetrics and Gynecology, IRCCS, San Raffaele Hospital, Milan, Italy
| | - F Puglisi
- Department of Medicine (DAME), University of Udine, Udine, Italy; Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano (PN), Italy
| | - S Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
9
|
Chan DW, Lam WY, Chen F, Yung MMH, Chan YS, Chan WS, He F, Liu SS, Chan KKL, Li B, Ngan HYS. Genome-wide DNA methylome analysis identifies methylation signatures associated with survival and drug resistance of ovarian cancers. Clin Epigenetics 2021; 13:142. [PMID: 34294135 PMCID: PMC8296615 DOI: 10.1186/s13148-021-01130-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In contrast to stable genetic events, epigenetic changes are highly plastic and play crucial roles in tumor evolution and development. Epithelial ovarian cancer (EOC) is a highly heterogeneous disease that is generally associated with poor prognosis and treatment failure. Profiling epigenome-wide DNA methylation status is therefore essential to better characterize the impact of epigenetic alterations on the heterogeneity of EOC. METHODS An epigenome-wide association study was conducted to evaluate global DNA methylation in a retrospective cohort of 80 mixed subtypes of primary ovarian cancers and 30 patients with high-grade serous ovarian carcinoma (HGSOC). Three demethylating agents, azacytidine, decitabine, and thioguanine, were tested their anti-cancer and anti-chemoresistant effects on HGSOC cells. RESULTS Global DNA hypermethylation was significantly associated with high-grade tumors, platinum resistance, and poor prognosis. We determined that 9313 differentially methylated probes (DMPs) were enriched in their relative gene regions of 4938 genes involved in small GTPases and were significantly correlated with the PI3K-AKT, MAPK, RAS, and WNT oncogenic pathways. On the other hand, global DNA hypermethylation was preferentially associated with recurrent HGSOC. A total of 2969 DMPs corresponding to 1471 genes were involved in olfactory transduction, and calcium and cAMP signaling. Co-treatment with demethylating agents showed significant growth retardation in ovarian cancer cells through differential inductions, such as cell apoptosis by azacytidine or G2/M cell cycle arrest by decitabine and thioguanine. Notably, azacytidine and decitabine, though not thioguanine, synergistically enhanced cisplatin-mediated cytotoxicity in HGSOC cells. CONCLUSIONS This study demonstrates the significant association of global hypermethylation with poor prognosis and drug resistance in high-grade EOC and highlights the potential of demethylating agents in cancer treatment.
Collapse
Affiliation(s)
- David W Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China.
| | - Wai-Yip Lam
- Lee's Pharmaceutical (HK) Ltd, 1/F Building 20E, Phase 3, Hong Kong Science Park, Shatin, Hong Kong, People's Republic of China
| | - Fushun Chen
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Mingo M H Yung
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Yau-Sang Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Wai-Sun Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Fangfang He
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Stephanie S Liu
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Karen K L Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Benjamin Li
- Lee's Pharmaceutical (HK) Ltd, 1/F Building 20E, Phase 3, Hong Kong Science Park, Shatin, Hong Kong, People's Republic of China
| | - Hextan Y S Ngan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China. .,Department of Obstetrics and Gynaecology, 6/F Professorial Block, Queen Mary Hospital, Pokfulam, Hong Kong, People's Republic of China.
| |
Collapse
|
10
|
Coughlan AY, Testa G. Exploiting epigenetic dependencies in ovarian cancer therapy. Int J Cancer 2021; 149:1732-1743. [PMID: 34213777 PMCID: PMC9292863 DOI: 10.1002/ijc.33727] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 01/02/2023]
Abstract
Ovarian cancer therapy has remained fundamentally unchanged for 50 years, with surgery and chemotherapy still the frontline treatments. Typically asymptomatic until advanced stages, ovarian cancer is known as “the silent killer.” Consequently, it has one of the worst 5‐year survival rates, as low as 30%. The most frequent driver mutations are found in well‐defined tumor suppressors, such as p53 and BRCA1/2. In recent years, it has become clear that, like the majority of other cancers, many epigenetic regulators are altered in ovarian cancer, including EZH2, SMARCA2/4 and ARID1A. Disruption of epigenetic regulators often leads to loss of transcriptional control, aberrant cell fate trajectories and disruption of senescence, apoptotic and proliferation pathways. These mitotically inherited epigenetic alterations are particularly promising targets for therapy as they are largely reversible. Consequently, many drugs targeting chromatin modifiers and other epigenetic regulators are at various stages of clinical trials for other cancers. Understanding the mechanisms by which ovarian cancer‐specific epigenetic processes are disrupted in patients can allow for informed targeting of epigenetic pathways tailored for each patient. In recent years, there have been groundbreaking new advances in disease modeling through ovarian cancer organoids; these models, alongside single‐cell transcriptomic and epigenomic technologies, allow the elucidation of the epigenetic pathways deregulated in ovarian cancer. As a result, ovarian cancer therapy may finally be ready to advance to next‐generation treatments. Here, we review the major developments in ovarian cancer, including genetics, model systems and technologies available for their study and the implications of applying epigenetic therapies to ovarian cancer.
Collapse
Affiliation(s)
- Aisling Y Coughlan
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Current update on malignant epithelial ovarian tumors. Abdom Radiol (NY) 2021; 46:2264-2280. [PMID: 34089360 DOI: 10.1007/s00261-021-03081-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/16/2023]
Abstract
Epithelial ovarian cancer (EOC) represents the most frequently occurring gynecological malignancy, accounting for more than 70% of ovarian cancer deaths. Preoperative imaging plays an important role in assessing the extent of disease and guides the next step in surgical decision-making and operative planning. In this article, we will review the multimodality imaging features of various subtypes of EOC. We will also discuss the role of imaging in the staging, management, and surveillance of EOC.
Collapse
|
12
|
Rumman M, Buck S, Polin L, Dzinic S, Boerner J, Winer IS. ONC201 induces the unfolded protein response (UPR) in high- and low-grade ovarian carcinoma cell lines and leads to cell death regardless of platinum sensitivity. Cancer Med 2021; 10:3373-3387. [PMID: 33932119 PMCID: PMC8124100 DOI: 10.1002/cam4.3858] [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: 10/06/2020] [Revised: 12/06/2020] [Accepted: 12/18/2020] [Indexed: 11/09/2022] Open
Abstract
Objectives Treatment of both platinum resistant high grade (HG) and low‐grade (LG) ovarian cancer (OVCA) poses significant challenges as neither respond well to conventional chemotherapy leading to morbidity and mortality. Identification of novel agents that can overcome chemoresistance is therefore critical. Previously, we have demonstrated that OVCA has basal upregulated unfolded protein response (UPR) and that targeting cellular processes leading to further and persistent upregulation of UPR leads to cell death. ONC201 is an orally bioavailable Dopamine Receptor D2 inhibitor demonstrating anticancer activity and was found to induce UPR. Given its unique properties, we hypothesized that ONC201 would overcome platinum resistance in OVCA. Methods Cisplatin sensitive and resistant HG OVCA and two primary LG OVCA cell lines were studied. Cell viability was determined using MTT assay. Cell migration was studied using wound healing assay. Apoptosis and mitochondrial membrane potential were investigated using flow cytometry. Analysis of pathway inhibition was performed by Western Blot. mRNA expression of UPR related genes were measured by qPCR. In vivo studies were completed utilizing axillary xenograft models. Co‐testing with conventional chemotherapy was performed to study synergy. Results ONC201 significantly inhibited cell viability and migration in a dose dependent manner with IC50’s from 1‐20 µM for both cisplatin sensitive and resistant HG and LG‐OVCA cell lines. ONC201 lead to upregulation of the pro‐apoptotic arm of the UPR, specifically ATF‐4/CHOP/ATF3 and increased the intrinsic apoptosispathway. The compensatory, pro‐survival PI3K/AKT/mTOR pathway was downregulated. In vivo, weekly dosing of single agent ONC201 decreased xenograft tumor size by ~50% compared to vehicle. ONC201 also demonstrated significant synergy with paclitaxel in a highly platinum resistant OVCA cell‐line (OV433). Conclusions Our findings demonstrate that ONC201 can effectively overcome chemoresistance in OVCA cells by blocking pro‐survival pathways and inducing the apoptotic arm of the UPR. This is a promising, orallybioavailable therapeutic agent to consider in clinical trials for patients with both HG and LG OVCA.
Collapse
Affiliation(s)
- Marufa Rumman
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Steven Buck
- Division of Hematology/Oncology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
| | - Lisa Polin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Sijana Dzinic
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Julie Boerner
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Ira S Winer
- Division of Gynecologic Oncology, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
13
|
Guo T, Dong X, Xie S, Zhang L, Zeng P, Zhang L. Cellular Mechanism of Gene Mutations and Potential Therapeutic Targets in Ovarian Cancer. Cancer Manag Res 2021; 13:3081-3100. [PMID: 33854378 PMCID: PMC8041604 DOI: 10.2147/cmar.s292992] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/19/2021] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is a common and complex malignancy with poor prognostic outcome. Most women with ovarian cancer are diagnosed with advanced stage disease due to a lack of effective detection strategies in the early stage. Traditional treatment with cytoreductive surgery and platinum-based combination chemotherapy has not significantly improved prognosis and 5-year survival rates are still extremely poor. Therefore, novel treatment strategies are needed to improve the treatment of ovarian cancer patients. Recent advances of next generation sequencing technologies have both confirmed previous known mutated genes and discovered novel candidate genes in ovarian cancer. In this review, we illustrate recent advances in identifying ovarian cancer gene mutations, including those of TP53, BRCA1/2, PIK3CA, and KRAS genes. In addition, we discuss advances in targeting therapies for ovarian cancer based on these mutated genes in ovarian cancer. Further, we associate between detection of mutation genes by liquid biopsy and the potential early diagnostic value in ovarian cancer.
Collapse
Affiliation(s)
- Tao Guo
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xue Dong
- Department of Gynecology, Cheng Du Shang Jin Nan Fu Hospital, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Shanli Xie
- First People's Hospital of Guangyuan, Guangyuan, Sichuan, 628000, People's Republic of China
| | - Ling Zhang
- Department of Gynecology and Obstetrics, Guangyuan Central Hospital, Guangyuan, Sichuan, 628000, People's Republic of China
| | - Peibin Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Lin Zhang
- Department of Forensic Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| |
Collapse
|
14
|
Dou L, Zhang Y. miR-4461 Regulates the Proliferation and Metastasis of Ovarian Cancer Cells and Cisplatin Resistance. Front Oncol 2021; 11:614035. [PMID: 33767986 PMCID: PMC7985457 DOI: 10.3389/fonc.2021.614035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
microRNAs (miRNAs) are of great significance in cancer treatment, which may have a desirable result on the regulation of tumorigenesis, progression, recurrence, and chemo-resistance of ovarian cancer. However, the research on the further potential application of miR-4461 in ovarian cancer is little and limited. Therefore, the study in this paper focus on the investigation of the of miR-4461 in ovarian cancer progression and chemo-resistance. The phenomenon that the proliferation and metastasis of ovarian cancer cells can be promoted by miR4461 is revealed in functional assays. Through the bioinformatics and luciferase reporter analysis, the PTEN is validated to be the direct target of miR-4461 in ovarian. The association between the expression of miR-4461 and PTEN is negative in in human ovarian cancer tissues. The distinction of growth and metastasis capacity between miR-4461 knockdown ovarian cancer cells and control cells is partially abolished by si-PTEN. Moreover, it was found that cisplatin treatment has obvious effect on the miR-4461 knockdown ovarian cancer cells. In summary, the data given in this paper indicate that the miR-4461 can be regarded as a potential onco-miRNA in ovarian cancer by targeting PTEN.
Collapse
Affiliation(s)
- Lei Dou
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yi Zhang
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
The mechanism of anticancer activity of the new synthesized compound - 6,7-Methylenedioxy-4-(2,4-dimethoxyphenyl)quinolin -2(1H)-one(12e) in human ovarian cancer cell lines. Taiwan J Obstet Gynecol 2021; 60:266-272. [PMID: 33678326 DOI: 10.1016/j.tjog.2021.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2020] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE Ovarian cancer is the most lethal of the gynecologic malignancies. Most women have advanced disease at diagnosis and require extensive debulking surgery and aggressive chemotherapy. Induction of apoptosis in cancer cells has been used as an important approach for cancer therapy. We examined the anticancer effect of 6,7-methylenedioxy-4-(2,4-dimethoxyphenyl)quinolin-2(1H)-one (12e) in human ovarian cancer cell line. MATERIALS AND METHODS The 6,7-methylenedioxy-4- (2,4-dimethoxyphenyl) quinolin-2 (1H) -one (12e) was synthesized and provided by Dr. Li-Jiau Huang of China Medical University. Cell viability analysis showed that 12e inhibited cell growth and induced cell death in time- and dose-dependent manners. In order to study the underlying cell death mechanism, 2774 and SKOV3 cells treated with 12e were studied by morphology, DAPI/TUNEL double staining, DNA gel electrophoresis. To search the mechanisms of anti-proliferative effect of 12e, cell cycle analysis was performed. Changes in proteins related to cell death were analyzed by Western blot. RESULTS 12e significantly induced apoptosis evidenced by morphological changes, TUNEL-DAPI double-staining and DNA fragmentation. Western blot analysis demonstrated that the protein level of Bcl-2 was decreased after treatment with 12e, while the level of p53 and Bax was increased. 12e treatment resulted in G2/M arrest through down modulation of cyclin B1 and cdk1. CONCLUSION These results suggested that 12e -induced growth inhibition was associated with cell cycle arrest and apoptosis.
Collapse
|
16
|
Gong H, Nie D, Li Z. Targeting Six Hallmarks of Cancer in Ovarian Cancer Therapy. Curr Cancer Drug Targets 2020; 20:853-867. [PMID: 32807056 DOI: 10.2174/1568009620999200816130218] [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: 05/21/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022]
Abstract
Normal cells must overcome multiple protective mechanisms to develop into cancer cells. Their new capabilities include self-sufficiency in growth signals and insensitivity to antigrowth signals, evasion of apoptosis, a limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis; these are also termed the six hallmarks of cancer. A deep understanding of the genetic and protein alterations involved in these processes has enabled the development of targeted therapeutic strategies and clinical trial design in the search for ovarian cancer treatments. Clinically, significantly longer progression-free survival has been observed in the single use of PARP, MEK, VEGF and Chk1/Chk2 inhibitors. However, the clinical efficacy of the targeted agents is still restricted to specific molecular subtypes and no trials illustrate a benefit in overall survival. Exploring novel drug targets or combining current feasible biological agents hold great promise to further improve outcomes in ovarian cancer. In this review, we intend to provide a comprehensive description of the molecular alterations involved in ovarian cancer carcinogenesis and of emerging biological agents and combined strategies that target aberrant pathways, which might shed light on future ovarian cancer treatment.
Collapse
Affiliation(s)
- Han Gong
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Nie
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Zhengyu Li
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
17
|
Brunetti M, Panagopoulos I, Kostolomov I, Davidson B, Heim S, Micci F. Mutation analysis and genomic imbalances of cells found in effusion fluids from patients with ovarian cancer. Oncol Lett 2020; 20:2273-2279. [PMID: 32782545 PMCID: PMC7400987 DOI: 10.3892/ol.2020.11782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/29/2020] [Indexed: 01/02/2023] Open
Abstract
Ovarian carcinomas and carcinosarcomas often cause malignant effusions, an accumulation within serous cavities of fluid containing cancer cells. Few studies have focused on the molecular alterations and genetic mechanisms behind effusion formation. The present study investigated the mutation status of TP53, PIK3CA, KRAS, HRAS, NRAS and BRAF in effusion fluids from 103 patients with ovarian cancer. In addition, array Comparative Genomic Hybridization (aCGH) analysis was performed on 20 effusions from patients with high-grade serous carcinoma (10 cases positive for TP53 mutation and 10 with TP53 wild-type). TP53 mutations, two of which were novel: c.826_830delCCTGT and c.475_476GC>TT, were identified in 44% of the cases. Mutations in KRAS, HRAS, and PIK3CA were identified in two, two and four cases, respectively. None of the effusions analysed showed NRAS or BRAF mutations. The aCGH analysis revealed highly imbalanced genomes similar to those described in primary ovarian carcinomas. No specific profile was indicated to distinguish tumors with TP53 mutations from those without. The molecular profiling of cells found in effusion fluids from patients with ovarian cancer thus showed considerable molecular heterogeneity. TP53 seems to be the most frequently mutated gene in these cells and may serve a leading role in the metastatic process.
Collapse
Affiliation(s)
- Marta Brunetti
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway.,Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0424 Oslo, Norway
| | - Ioannis Panagopoulos
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway
| | - Ilyá Kostolomov
- Section for Applied Informatics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway
| | - Ben Davidson
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0424 Oslo, Norway
| | - Sverre Heim
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0424 Oslo, Norway
| | - Francesca Micci
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
18
|
Ge J, Han T, Shan L, Na J, Li Y, Wang J. Long non-coding RNA THOR promotes ovarian Cancer cells progression via IL-6/STAT3 pathway. J Ovarian Res 2020; 13:72. [PMID: 32552789 PMCID: PMC7302152 DOI: 10.1186/s13048-020-00672-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
Background Ovarian cancer (OC) is one of the most common malignant tumors in the world. The prognosis of OC remains poor due to the advanced stage and distant metastasis at the time of diagnosis. Recently, a novel lncRNA, THOR (testis-associated highly conserved oncogenic long non-coding RNA), was characterized in human cancers and shown to exhibit an oncogenic role. However, the role of THOR in OC remains unclear. Methods RT-PCR and western blot analysis were used to detect the expression of THOR, p-STAT3 and IL-6. The impact of THOR on OC proliferation, metastasis and self-renewal was investigated in vitro and in vivo. The prognostic value of THOR was determined in OC patient cohorts. Results In this study, our results find that THOR is markedly upregulated in human OC tissues and predicts the poor prognosis of OC patients. Functional studies have revealed that knockdown of THOR inhibits the growth, metastasis and self-renewal of OC cells. Mechanistically, THOR drives OC cell progression via the IL-6/STAT3 signaling. Moreover, the specific STAT3 inhibitor S3I-201 or IL-6R inhibitor tocilizumab diminish the discrepancy in the growth, metastatic and self-renewal capacity between THOR-silenced OC cells and control cells, which further confirm that IL-6/STAT3 is required in THOR-driven OC cells progression. Conclusion Our findings reveal that THOR could promote OC cells growth, metastasis and self-renewal by activating IL-6/STAT3 signaling and may be a good predictive factor and therapeutic target.
Collapse
Affiliation(s)
- Jing Ge
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, China.,Department of Gynecology and Obstetrics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning Province, China
| | - Tao Han
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, China.,Department of Oncology, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning Province, China
| | - Lili Shan
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, China.,Department of Gynecology and Obstetrics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning Province, China
| | - Jing Na
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, China.,Department of Gynecology and Obstetrics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning Province, China
| | - Ya Li
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, China.,Department of Gynecology and Obstetrics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning Province, China
| | - Jun Wang
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, Liaoning Province, China. .,Department of Gynecology and Obstetrics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning Province, China.
| |
Collapse
|
19
|
EMR 20006-012: A phase II randomized double-blind placebo controlled trial comparing the combination of pimasertib (MEK inhibitor) with SAR245409 (PI3K inhibitor) to pimasertib alone in patients with previously treated unresectable borderline or low grade ovarian cancer. Gynecol Oncol 2019; 156:301-307. [PMID: 31870556 DOI: 10.1016/j.ygyno.2019.12.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To compare the combination of a MEK inhibitor (pimasertib) and a PI3K inhibitor (SAR245409) to pimasertib alone in recurrent unresectable borderline/low malignant potential (LMP) or low-grade serous ovarian carcinoma (LGSOC), determining whether combination is superior. METHODS Patients with previously treated, recurrent LMP or LGSOC with measurable disease received either combination of pimasertib (60 mg daily) + SAR245409 (SAR) (70 mg daily) or pimasertib alone (60 mg BID) until progression or unacceptable toxicity. Primary endpoint was objective response rate (ORR) by RECIST 1.1, determining whether combination was superior to pimasertib alone. Secondary endpoints included progression free survival (PFS), disease control, and adverse events. RESULTS Sixty-five patients were randomized between September 2012 and December 2014. ORR was 9.4% (80% CI, 3.5 to 19.7) in the combination arm and 12.1% (80% CI, 5.4 to 22.8) in the pimasertib alone arm. Median PFS was 7.23 months (80% CI, 5.06 to -) and 9.99 (80% CI, 7.39 to 10.35) for pimasertib alone and pimasertib + SAR, respectively. Six-month PFS was 63.5% (80% CI, 47.2% to 75.9%) and 70.8% (80% CI, 56.9% to 80.9%). Eighteen (56.3%) patients in the combination arm and 19 (57.6%) patients in the pimasertib alone arm discontinued the trial. The study was terminated early because of low ORR and high rate of discontinuation. CONCLUSIONS Response to pimasertib alone (ORR 12%) suggests that MEK inhibition could be used as an alternative treatment method to cytotoxic chemotherapy in this population. The MEK inhibitor alone was as effective as the combination, although the trial was limited by small numbers. Additional studies investigating the role of single agent or combination MEK and PI3K inhibition are warranted to further evaluate the utility of these treatments and describe a standard of care for LGSOC.
Collapse
|
20
|
Zhao W, Han T, Li B, Ma Q, Yang P, Li H. miR-552 promotes ovarian cancer progression by regulating PTEN pathway. J Ovarian Res 2019; 12:121. [PMID: 31815639 PMCID: PMC6900846 DOI: 10.1186/s13048-019-0589-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 11/04/2019] [Indexed: 12/29/2022] Open
Abstract
Background Increasing researches have demonstrated the critical functions of MicroRNAs (miRNAs) in the progression of malignant tumors, including ovarian cancer. It was reported that miR-552 was an important oncogene in both breast cancer and colorectal cancer. However, the role of miR-552 in ovarian cancer (OC) remains to be elucidated. Methods RT-PCR and western blot analysis were used to detect the expression of miR-552 and PTEN. The impact of miR-552 on ovarian cancer proliferation and metastasis was investigated in vitro. The prognostic value of miR-552 was evaluated using the online bioinformatics tool Kaplan-Meier plotter. Results In the present study, we for first found that miR-552 was upregulated in ovarian cancer, especially in metastatic and recurrence ovarian cancer. Forced miR-552 expression promotes the growth and metastasis of ovarian cancer cells. Consistently, miR-552 interference inhibits the proliferation and metastasis of ovarian cancer cells. Mechanically, bioinformatics and luciferase reporter analysis identified Phosphatase and tension homolog (PTEN) as a direct target of miR-552. miR-552 downregulated the PTEN mRNA and protein expression in ovarian cancer cells. Furthermore, the PTEN siRNA abolishes the discrepancy of growth and metastasis capacity between miR-552 mimic ovarian cells and control cells. More importantly, upregulation of miR-552 predicts the poor prognosis of ovarian cancer patients. Conclusion Our findings revealed that miR-552 could promote ovarian cancer cells progression by targeting PTEN signaling and might therefore be useful to predict patient prognosis.
Collapse
Affiliation(s)
- Wenman Zhao
- Department of General surgery, Cao county people's hospital, East of Qinghe Road, Heze, 274400, Shandong province, China.
| | - Tao Han
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning Province, China
| | - Bao Li
- Department of General surgery, Cao county people's hospital, East of Qinghe Road, Heze, 274400, Shandong province, China
| | - Qianyun Ma
- Department of Urology surgery, First Affiliated Hospital of Second Military Medical University, Shanghai, 200433, China
| | - Pinghua Yang
- Department of Biliary Tract Surgery, Third Affiliated Hospital of Second Military Medical University, Shanghai, 200438, China.
| | - Hengyu Li
- Department of Breast and Thyroid surgery, First Affiliated Hospital of Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
21
|
Voutsadakis IA. Low-grade serous ovarian carcinoma: an evolution toward targeted therapy. Int J Gynecol Cancer 2019; 30:1619-1626. [PMID: 31780569 DOI: 10.1136/ijgc-2019-000832] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/16/2019] [Accepted: 09/24/2019] [Indexed: 11/04/2022] Open
Abstract
Low-grade serous ovarian carcinoma and its high-grade serous ovarian carcinoma counterpart differ in their precursor lesions, molecular profile, natural history, and response to therapies. As such, low-grade serous ovarian carcinoma needs to be studied separately from high-grade serous ovarian carcinoma, despite challenges stemming from its rarity. A deeper understanding of the pathogenesis of low-grade serous ovarian carcinoma and the most common molecular defects and pathways involved in the carcinogenesis of the ovarian epithelium from normal to serous borderline ovarian tumors to low-grade serous ovarian carcinoma will help develop better therapies. By adopting targeted approaches there may be an opportunity to integrate novel therapies without the need for robust numbers in clinical trials. This manuscript will discuss low-grade serous ovarian carcinoma and focus on the arising treatments being developed with an improved understanding of the pathogenesis of this disease.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Section of Internal Medicine Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
22
|
Yu HL, Ma XD, Tong JF, Li JQ, Guan XJ, Yang JH. WTAP is a prognostic marker of high-grade serous ovarian cancer and regulates the progression of ovarian cancer cells. Onco Targets Ther 2019; 12:6191-6201. [PMID: 31496724 PMCID: PMC6689666 DOI: 10.2147/ott.s205730] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/27/2019] [Indexed: 01/09/2023] Open
Abstract
Background The Wilms' tumor suppressor WT1 is reported to work in a range of physiological processes at both transcriptional and posttranscriptional level. WT1-associating protein (WTAP), a nuclear protein co-localized with splicing factors, also plays a vital role in cellular function and cancer progression. However, little is known about the role of WTAP in ovarian cancer and the underlying mechanism. Materials and methods To evaluate the expression of WTAP, multiple means were applied in clinical tissues, including immunohistochemistry, quantitative reverse transcriptase PCR (qRT-PCR), and Western blot. Two representative ovarian cancer cell lines (3AO and SKOV3) were used to assess the malignant influence of WTAP on proliferation, apoptosis, and migration. To explore its function, WTAP was additionally down-regulated by lentivirus. Results High expression of WTAP in high-grade serous ovarian carcinoma (HGSOC) predicted a shorter overall survival (P<0.01). Furthermore, WTAP expression was higher in HGSOC, compared with that in normal ovary group (P<0.01), benign ovarian tumor group (P<0.01), and non-HGSOC group (P<0.05). In HGSOC, high expression of WTAP was significantly related with the lymph node metastasis (P<0.05). In ovarian cancer cell lines, cell proliferation and migration were considerably reduced after WTAP was down-regulated, while apoptotic rate was increased. Moreover, the effect of WTAP in 3AO and SKOV3 might be relevant with MAPK and AKT signaling pathways. Conclusion WTAP is highly expressed in HGSOC, and indicates a worse survival outcome. Therefore, it is highly possible that WTAP has a prognostic implication in the patients of HGSOC. In addition, WTAP down-regulation also plays a tumor suppressor role in 3AO and SKOV3 cell lines.
Collapse
Affiliation(s)
- Hai-Lan Yu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xu-Dong Ma
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jin-Fei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jian-Qiong Li
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xiao-Jing Guan
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jian-Hua Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
23
|
Dual-drug loaded micelle for combinatorial therapy targeting HIF and mTOR signaling pathways for ovarian cancer treatment. J Control Release 2019; 307:272-281. [PMID: 31260753 DOI: 10.1016/j.jconrel.2019.06.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/20/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022]
Abstract
Mutations in the tumor protein (TP53) and the mammalian target of rapamycin (mTOR) pathway have been elucidated as driver mutations in ovarian carcinomas that transform into an invasive phenotype under hypoxic conditions. Chetomin (CHE) targets the hypoxic pathway while Everolimus (EVR) acts on the mTOR pathway. Poor aqueous solubilities of both compounds limit their clinical applications. Diblock copolymer nanoplatforms of methoxy poly(ethylene glycol)2000-block-poly (lactic acid)1800 (mPEG2000-b-PLA1800) and (mPEG4000-b-PLA2200) were used to formulate individual and dual drug loaded micelles (DDM) using the solvent evaporation method. The CHE micelles (CHE-M) had a size of 21 nm with CHE loading of 0.5 mg/mL while the EVR micelles (EVR-M) and the DDM had a size around 35 and 39 nm, respectively, with EVR loading up to 2.3 mg/mL. The anti-proliferative effects of these micelles have been tested in vitro in three ovarian cell lines (ES2, OVCAR3 and TOV21G) with the DDM exhibiting a strong synergistic anti-proliferative effect in the ES2 and the TOV21G cells. The DDM were able to significantly induce tumor regression in ES2 ovarian xenograft mouse models by inhibiting angiogenesis and inducing apoptosis when compared to the individual micelles. The inhibition of hypoxia inducible factor (HIF) and the mTOR pathways has been elucidated using immunohistochemistry studies. In conclusion, we have developed a mPEG-b-PLA based micellar nanoplatform that could prevent drug resistance by delivering multiple drugs at therapeutically relevant concentrations for effectively treating ovarian carcinomas.
Collapse
|
24
|
Iavarone C, Zervantonakis IK, Selfors LM, Palakurthi S, Liu JF, Drapkin R, Matulonis UA, Hallberg D, Velculescu VE, Leverson JD, Sampath D, Mills GB, Brugge JS. Combined MEK and BCL-2/X L Inhibition Is Effective in High-Grade Serous Ovarian Cancer Patient-Derived Xenograft Models and BIM Levels Are Predictive of Responsiveness. Mol Cancer Ther 2019; 18:642-655. [PMID: 30679390 PMCID: PMC6399746 DOI: 10.1158/1535-7163.mct-18-0413] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/30/2018] [Accepted: 01/14/2019] [Indexed: 11/16/2022]
Abstract
Most patients with late-stage high-grade serous ovarian cancer (HGSOC) initially respond to chemotherapy but inevitably relapse and develop resistance, highlighting the need for novel therapies to improve patient outcomes. The MEK/ERK pathway is activated in a large subset of HGSOC, making it an attractive therapeutic target. Here, we systematically evaluated the extent of MEK/ERK pathway activation and efficacy of pathway inhibition in a large panel of well-annotated HGSOC patient-derived xenograft models. The vast majority of models were nonresponsive to the MEK inhibitor cobimetinib (GDC-0973) despite effective pathway inhibition. Proteomic analyses of adaptive responses to GDC-0973 revealed that GDC-0973 upregulated the proapoptotic protein BIM, thus priming the cells for apoptosis regulated by BCL2-family proteins. Indeed, combination of both MEK inhibitor and dual BCL-2/XL inhibitor (ABT-263) significantly reduced cell number, increased cell death, and displayed synergy in vitro in most models. In vivo, GDC-0973 and ABT-263 combination was well tolerated and resulted in greater tumor growth inhibition than single agents. Detailed proteomic and correlation analyses identified two subsets of responsive models-those with high BIM at baseline that was increased with MEK inhibition and those with low basal BIM and high pERK levels. Models with low BIM and low pERK were nonresponsive. Our findings demonstrate that combined MEK and BCL-2/XL inhibition has therapeutic activity in HGSOC models and provide a mechanistic rationale for the clinical evaluation of this drug combination as well as the assessment of the extent to which BIM and/or pERK levels predict drug combination effectiveness in chemoresistant HGSOC.
Collapse
Affiliation(s)
- Claudia Iavarone
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Ioannis K Zervantonakis
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Laura M Selfors
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Sangeetha Palakurthi
- Belfer Institute for Applied Cancer Res, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joyce F Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ronny Drapkin
- Penn Ovarian Cancer Res Center, Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Dorothy Hallberg
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Deepak Sampath
- Translational Oncology, Genentech, South San Francisco, California
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joan S Brugge
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
25
|
Elsherif S, Javadi S, Viswanathan C, Faria S, Bhosale P. Low-grade epithelial ovarian cancer: what a radiologist should know. Br J Radiol 2019; 92:20180571. [PMID: 30604635 DOI: 10.1259/bjr.20180571] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ovarian cancer accounts for the death of over 100,000 females every year and is the most lethal gynecological malignancy. Low-grade serous ovarian carcinoma (LGSOC) and high-grade serous ovarian carcinoma (HGSOC) have been found to represent two distinct entities based on their molecular differences, clinical course, and response to chemotherapy. Currently, all ovarian cancers are staged according to the revised staging system of the International Federation of Gynecology and Obstetrics (FIGO). Imaging plays an integral role in the diagnosis, staging, and follow-up of ovarian cancers. This review will be based on the two-tier grading system of epithelial ovarian cancers, with the main emphasis on serous ovarian cancer, and the role of imaging to characterize low-grade vs high-grade tumors and monitor disease recurrence during follow-up.
Collapse
Affiliation(s)
- Sherif Elsherif
- 1 Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Sanaz Javadi
- 1 Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Chitra Viswanathan
- 1 Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Silvana Faria
- 1 Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Priya Bhosale
- 1 Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| |
Collapse
|
26
|
Klein O, Kanter F, Kulbe H, Jank P, Denkert C, Nebrich G, Schmitt WD, Wu Z, Kunze CA, Sehouli J, Darb‐Esfahani S, Braicu I, Lellmann J, Thiele H, Taube ET. MALDI‐Imaging for Classification of Epithelial Ovarian Cancer Histotypes from a Tissue Microarray Using Machine Learning Methods. Proteomics Clin Appl 2018; 13:e1700181. [DOI: 10.1002/prca.201700181] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 10/31/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Oliver Klein
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Berlin‐Brandenburg Center for Regenerative TherapiesCharité—Universitätsmedizin Berlin 13353 Berlin Germany
| | - Frederic Kanter
- Institute of Mathematics and Image ComputingUniversität zu Lübeck Lübeck Germany
| | - Hagen Kulbe
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Department of GynecologyCharité—Universitätsmedizin Berlin 13353 Berlin Germany
- Fraunhofer—Institute for Medical Image Computing MEVIS 23562 Lübeck Germany
| | - Paul Jank
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Institute of PathologyCharité—Universitätsmedizin Berlin 10117 Berlin Germany
| | - Carsten Denkert
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Institute of PathologyCharité—Universitätsmedizin Berlin 10117 Berlin Germany
| | - Grit Nebrich
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Berlin‐Brandenburg Center for Regenerative TherapiesCharité—Universitätsmedizin Berlin 13353 Berlin Germany
| | - Wolfgang D. Schmitt
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Institute of PathologyCharité—Universitätsmedizin Berlin 10117 Berlin Germany
| | - Zhiyang Wu
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Berlin‐Brandenburg Center for Regenerative TherapiesCharité—Universitätsmedizin Berlin 13353 Berlin Germany
| | - Catarina A. Kunze
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Institute of PathologyCharité—Universitätsmedizin Berlin 10117 Berlin Germany
| | - Jalid Sehouli
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Department of GynecologyCharité—Universitätsmedizin Berlin 13353 Berlin Germany
- Fraunhofer—Institute for Medical Image Computing MEVIS 23562 Lübeck Germany
| | - Silvia Darb‐Esfahani
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Institute of Pathology Spandau 13589 Berlin Germany
| | - Ioana Braicu
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Department of GynecologyCharité—Universitätsmedizin Berlin 13353 Berlin Germany
- Fraunhofer—Institute for Medical Image Computing MEVIS 23562 Lübeck Germany
| | - Jan Lellmann
- Institute of Mathematics and Image ComputingUniversität zu Lübeck Lübeck Germany
| | - Herbert Thiele
- Fraunhofer—Institute for Medical Image Computing MEVIS 23562 Lübeck Germany
| | - Eliane T. Taube
- Charité—Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
- Institute of PathologyCharité—Universitätsmedizin Berlin 10117 Berlin Germany
| |
Collapse
|
27
|
Prognostic significance of Ki-67 levels and hormone receptor expression in low-grade serous ovarian carcinoma: an investigation of the Tumor Bank Ovarian Cancer Network. Hum Pathol 2018; 85:299-308. [PMID: 30428389 DOI: 10.1016/j.humpath.2018.10.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/18/2018] [Accepted: 10/22/2018] [Indexed: 12/25/2022]
Abstract
Low-grade serous ovarian carcinoma (LGSOC) has recently come up as a distinct rare entity of epithelial ovarian cancer. Predictive and prognostic markers are not well studied yet. Because Ki-67 and hormone receptors (HR) have been established as relevant cancer biomarkers in several malignant tumors, we evaluated Ki-67 and HR expression rates by immunohistochemistry in 68 patients with LGSOC. We used a standardized cutoff finder algorithm to analyze prognostic significance for overall survival (OS) and progression-free survival (PFS). Cox regression showed a significant continuous decrease in OS for higher proliferation rates with an HR of 1.07% (95% confidence interval, 1.01%-3.67%; P = .048) but not in PFS (P = .86). Cutoff finder analysis revealed the best possible cutoff for OS at 6.28% (P = .04) and for PFS at 1.85% proliferative activity (P = .04). Estrogen receptors (ERs) were expressed in most LGSOC patients (n = 61; 89.7%), progesterone receptor (PR) in about half of patients (n = 33; 48.5%). For both ER/PR, a statistically significant cutoff for PFS could be determined, which was at 75% of positive tumor cells for ER (P = .02) and at 15% of positive tumor cells for PR (P = .03). For OS, HR expression showed a tendency toward better OS for HR-positive tumors but did not turn out statistically significant. Our results show that Ki-67 is a valuable prognostic marker in the subgroup of LGSOC. We could also show that most LGSOCs express HRs but that this expression is associated with a better PFS, a finding valuable in times of antihormonal therapy in LGSOC.
Collapse
|
28
|
Shih AJ, Menzin A, Whyte J, Lovecchio J, Liew A, Khalili H, Bhuiya T, Gregersen PK, Lee AT. Identification of grade and origin specific cell populations in serous epithelial ovarian cancer by single cell RNA-seq. PLoS One 2018; 13:e0206785. [PMID: 30383866 PMCID: PMC6211742 DOI: 10.1371/journal.pone.0206785] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/18/2018] [Indexed: 12/15/2022] Open
Abstract
Here we investigated different cell populations within ovarian cancer using single-cell RNA seq: fourteen samples from nine patients with differing grades (high grade, low grade and benign) as well as different origin sites (primary and metastatic tumor site, ovarian in origin and fallopian in origin). We were able to identify sixteen distinct cell populations with specific cells correlated to high grade tumors, low grade tumors, benign and one population unique to a patient with a breast cancer relapse. Furthermore the proportion of these populations changes from primary to metastatic in a shift from mainly epithelial cells to leukocytes with few cancer epithelial cells in the metastases. Differential gene expression shows myeloid lineage cells are the primary cell group expressing soluble factors in primary samples while fibroblasts do so in metastatic samples. The leukocytes that were captured did not seem to be suppressed through known pro-tumor cytokines from any of the cell populations. Single cell RNA-seq is necessary to de-tangle cellular heterogeneity for better understanding of ovarian cancer progression.
Collapse
Affiliation(s)
- Andrew J. Shih
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- * E-mail:
| | - Andrew Menzin
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, United States of America
| | - Jill Whyte
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, United States of America
| | - John Lovecchio
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, United States of America
| | - Anthony Liew
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Houman Khalili
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Tawfiqul Bhuiya
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, United States of America
| | - Peter K. Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, United States of America
| | - Annette T. Lee
- Robert S. Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, United States of America
| |
Collapse
|
29
|
Zhang W, Wang L, Xin Z. Combination of serum CA19-9 and CA125 levels and contrast-enhanced ultrasound parametric data facilitates to differentiate ovarian serous carcinoma from ovarian malignant epithelial cancer. Medicine (Baltimore) 2018; 97:e0358. [PMID: 29668586 PMCID: PMC5916679 DOI: 10.1097/md.0000000000010358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ovarian serous carcinoma (OSC) is semimalignant ovarian tumors that localized in the ovary at the initial presentation, and can be surgically resected in an excellent prognosis. In contrast, surgical treatment plus adjuvant chemotherapy for ovarian malignant epithelial cancer (OMEC) is the standard treatment stratagem that is painful with poor prognosis and cost quite expensively. Thus, the accurate preoperative differentiation of OSC from OMEC is important for determining the treatment methods and decreasing the cost for individual patients. This study aims to determine whether contrast enhanced ultrasound (CEUS) together with CA19-9/CA125 can improve the ability to differentiate ovarian serous carcinoma from ovarian malignant epithelial cancer. The positive rate of cancer antigen (CA) 199 and CA125 in ovarian malignant epithelial tumors was 68% and 94%, respectively, which was a higher incidence than in the serous carcinoma. The mean serum CA19-9 and CA125 concentration was significantly higher in the patients with ovarian malignant epithelial tumors (CA19-9, 514.0 ± 104.8 U/mL; CA125, 440.0 ± 154.8 U/mL) than that in the patients with ovarian serous carcinoma (CA19-9, 58.0 ± 14.3 U/mL; CA125, 63.0 ± 25.8 U/mL). The time to peak in ovarian serous carcinoma was significantly longer than in ovarian malignant epithelial tumors. However, the peak intensity, area under the curve, and washout time in ovarian serous carcinoma were significantly lower than in ovarian malignant epithelial tumors. The addition of CA19-9/CA125 increased the sensitivities from 79% CEUS only to 85% for CEUS plus CA19-9/CA125 data, and increased the specificities from 65% CEUS only to 91% for CEUS plus the CA19-9/CA125 information. Thus, the addition of the serum CA19-9/CA125 levels to parametric CEUS data was of significant diagnostic value for differentiating OSC from OMEC.
Collapse
|
30
|
Low-grade serous carcinoma (mullerian/ovarian type) of the paratestis presenting as diffuse metastatic disease of unknown origin: Case report of an uncommon tumor with an unusual presentation. HUMAN PATHOLOGY: CASE REPORTS 2018. [DOI: 10.1016/j.ehpc.2017.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
31
|
Ciucci A, Zannoni GF, Buttarelli M, Martinelli E, Mascilini F, Petrillo M, Ferrandina G, Scambia G, Gallo D. Ovarian low and high grade serous carcinomas: hidden divergent features in the tumor microenvironment. Oncotarget 2018; 7:68033-68043. [PMID: 27462782 PMCID: PMC5356537 DOI: 10.18632/oncotarget.10797] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/13/2016] [Indexed: 01/22/2023] Open
Abstract
Only recently low-grade serous carcinoma (LGSOC) of the ovary has been recognized as a disease entity distinct from the more common high-grade serous carcinoma (HGSOC), with significant differences in pathogenesis and clinical and pathologic features. The present study aimed at evaluating whether the different natural histories and patterns of response to therapy demonstrated for LGSOC and HGSOC, along with a diverse genomic landscape, may also reside in the supporting tumor stroma, specifically in the state of differentiation and activation of tumor associated macrophages (TAMs). TAMs play complex roles in tumorigenesis since they are believed to possess both tumor rejecting (M1 macrophages) and tumor promoting (M2 macrophages) activities. Here we showed that, when compared to HGSOC (n = 55), LGSOC patients (n = 25) exhibited lower density of tumor-infiltrating CD68+ macrophage, along with an attenuated M2-skewed (CD163+) phenotype. Accordingly, assessment of intratumoral vascularization and of matrix metalloproteinase 9 expression (a key protein involved in tumor invasion and metastasis) revealed lower expression in LGSOC compared to HGSOC patients, in line with emerging evidence supporting a role for TAMs in all aspects of tumor initiation, growth, and development. In conclusion, results from the present study demonstrate that microenvironmental factors contribute greatly to determine clinical and pathological features that differentiate low and high grade serous ovarian carcinomas. This understanding may increase possibilities and opportunities to improve disease control and design new therapeutic strategies.
Collapse
Affiliation(s)
- Alessandra Ciucci
- Unit of Translational Medicine for Women and Children Health, Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Gian Franco Zannoni
- Department of Pathology, Catholic University of the Sacred Heart, Rome, Italy
| | - Marianna Buttarelli
- Unit of Translational Medicine for Women and Children Health, Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Enrica Martinelli
- Unit of Translational Medicine for Women and Children Health, Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Floriana Mascilini
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Marco Petrillo
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Gabriella Ferrandina
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy.,Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Giovanni Scambia
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - Daniela Gallo
- Unit of Translational Medicine for Women and Children Health, Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
32
|
Jang JYA, Yanaihara N, Pujade-Lauraine E, Mikami Y, Oda K, Bookman M, Ledermann J, Shimada M, Kiyokawa T, Kim BG, Matsumura N, Kaku T, Kuroda T, Nagayoshi Y, Kawabata A, Iida Y, Kim JW, Quinn M, Okamoto A. Update on rare epithelial ovarian cancers: based on the Rare Ovarian Tumors Young Investigator Conference. J Gynecol Oncol 2018; 28:e54. [PMID: 28541641 PMCID: PMC5447152 DOI: 10.3802/jgo.2017.28.e54] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/17/2017] [Accepted: 04/18/2017] [Indexed: 02/08/2023] Open
Abstract
There has been significant progress in the understanding of the pathology and molecular biology of rare ovarian cancers, which has helped both diagnosis and treatment. This paper provides an update on recent advances in the knowledge and treatment of rare ovarian cancers and identifies gaps that need to be addressed by further clinical research. The topics covered include: low-grade serous, mucinous, and clear cell carcinomas of the ovary. Given the molecular heterogeneity and the histopathological rarity of these ovarian cancers, the importance of designing adequately powered trials or finding statistically innovative ways to approach the treatment of these rare tumors has been emphasized. This paper is based on the Rare Ovarian Tumors Conference for Young Investigators which was presented in Tokyo 2015 prior to the 5th Ovarian Cancer Consensus Conference of the Gynecologic Cancer InterGroup (GCIG).
Collapse
Affiliation(s)
- Ji Yon Agnes Jang
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Nozomu Yanaihara
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan.
| | - Eric Pujade-Lauraine
- Unité Cancer de la Femme et Recherche Clinique, Hôpitaux Universitaires Paris Centre, site Hôtel-Dieu, Université Paris Descartes, Paris, France
| | - Yoshiki Mikami
- Department of Diagnostic Pathology, Kumamoto University Hospital, Kumamoto, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | - Muneaki Shimada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Japan
| | - Takako Kiyokawa
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Byoung Gie Kim
- Gynecologic Cancer Center, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kyoto University, Kyoto, Japan
| | - Tsunehisa Kaku
- Department of Health Sciences, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Takafumi Kuroda
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoko Nagayoshi
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Ayako Kawabata
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasushi Iida
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Jae Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, Korea
| | - Michael Quinn
- Women's Cancer Research Center, Royal Women's Hospital, Melbourne, Australia
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
33
|
Qiu C, Lu N, Wang X, Zhang Q, Yuan C, Yan S, Dongol S, Li Y, Sun X, Sun C, Zhang Z, Zheng W, Kong B. Gene expression profiles of ovarian low-grade serous carcinoma resemble those of fallopian tube epithelium. Gynecol Oncol 2017; 147:634-641. [DOI: 10.1016/j.ygyno.2017.09.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 12/20/2022]
|
34
|
Moro F, Baima Poma C, Zannoni GF, Vidal Urbinati A, Pasciuto T, Ludovisi M, Moruzzi MC, Carinelli S, Franchi D, Scambia G, Testa AC. Imaging in gynecological disease (12): clinical and ultrasound features of invasive and non-invasive malignant serous ovarian tumors. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2017; 50:788-799. [PMID: 28101917 DOI: 10.1002/uog.17414] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 06/06/2023]
Abstract
OBJECTIVES To describe clinical and ultrasound features of different subclasses of malignant serous ovarian tumors according to the World Health Organization 2014 classification. METHODS Patients with a histological diagnosis of borderline tumor (BOT), non-invasive and invasive low-grade serous carcinoma (LGSC) and high-grade serous carcinoma (HGSC), who had undergone preoperative ultrasound examination, were retrospectively identified from two ultrasound centers. The masses were described using the terms of the International Ovarian Tumor Analysis Group. RESULTS Sixty-four (15.8%) women had a serous BOT, 11 (2.7%) a non-invasive LGSC, 31 (7.6%) an invasive LGSC and 300 (73.9%) had a HGSC. The vast majority of BOTs (82.3%) and non-invasive LGSCs (90.9%) were Stage I according to the International Federation of Gynecology and Obstetrics (FIGO) classification scheme, whereas most invasive LGSCs (74.2%) and HGSCs (74.0%) were FIGO Stage III. On ultrasound examination, most borderline lesions were described as unilocular-solid (54.7%) or as multilocular-solid (29.7%) cysts. Papillary projections were present in 52 (81.3%) BOTs. Most non-invasive LGSCs (63.6%) were multilocular-solid cysts and 81.8% had papillary projections. Invasive LGSCs were multilocular-solid cysts in 54.8% of cases, and papillary projections were present in 32.3% of lesions. HGSCs were multilocular-solid (32.7%) or solid (64.0%) masses, with papillary projections in only 7% of cases. CONCLUSIONS Papillary projections were the most typical ultrasound feature of non-invasive (borderline and low-grade) malignant serous tumors, while the presence of solid components but few, if any, papillations was the most representative feature of invasive (low-grade and high-grade) serous tumors. Copyright © 2017 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- F Moro
- Department of Women's and Children's Health, Catholic University of the Sacred Heart, Rome, Italy
| | - C Baima Poma
- Department of Women's and Children's Health, Catholic University of the Sacred Heart, Rome, Italy
| | - G F Zannoni
- Institute of Histopathology, Catholic University of the Sacred Heart, Rome, Italy
| | - A Vidal Urbinati
- Preventive Gynecology Unit, Division of Gynecology, European Institute of Oncology, Milan, Italy
| | - T Pasciuto
- Department of Women's and Children's Health, Catholic University of the Sacred Heart, Rome, Italy
| | - M Ludovisi
- Department of Women's and Children's Health, Catholic University of the Sacred Heart, Rome, Italy
| | - M C Moruzzi
- Department of Women's and Children's Health, Catholic University of the Sacred Heart, Rome, Italy
| | - S Carinelli
- Institute of Histopathology, European Institute of Oncology, Milan, Italy
| | - D Franchi
- Preventive Gynecology Unit, Division of Gynecology, European Institute of Oncology, Milan, Italy
| | - G Scambia
- Department of Women's and Children's Health, Catholic University of the Sacred Heart, Rome, Italy
| | - A C Testa
- Department of Women's and Children's Health, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
35
|
da Silva RF, Cardozo DM, Rodrigues GOL, Souza-Araújo CND, Migita NA, Andrade LALDA, Derchain S, Yunes JA, Guimarães F. CAISMOV24, a new human low-grade serous ovarian carcinoma cell line. BMC Cancer 2017; 17:756. [PMID: 29132324 PMCID: PMC5683553 DOI: 10.1186/s12885-017-3716-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 10/30/2017] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The spontaneous immortalization of primary malignant cells is frequently assigned to their genetic instability during in vitro culturing. In this study, the new epithelial ovarian cancer cell line CAISMOV24 was described and compared with its original low-grade serous ovarian carcinoma. METHODS The in vitro culture was established with cells isolated from ascites of a 60-year-old female patient with recurrent ovarian cancer. The CAISMOV24 line was assessed for cell growth, production of soluble biomarkers, expression of surface molecules and screened for typical mutations found in serous ovarian carcinoma. Additionally, comparative genomic hybridization was employed to compare genomic alterations between the CAISMOV24 cell line and its primary malignant cells. RESULTS CAISMOV24 has been in continuous culture for more than 30 months and more than 100 in vitro passages. The cell surface molecules EpCAM, PVR and CD73 are overexpressed on CAISMOV24 cells compared to the primary malignant cells. CAISMOV24 continues to produce CA125 and HE4 in vitro. Although the cell line had developed alongside the accumulation of genomic alterations (28 CNV in primary cells and 37 CNV in CAISMOV24), most of them were related to CNVs already present in primary malignant cells. CAISMOV24 cell line harbored KRAS mutation with wild type TP53, therefore it is characterized as low-grade serous carcinoma. CONCLUSION Our results corroborate with the idea that genomic alterations, depicted by CNVs, can be used for subtyping epithelial ovarian carcinomas. Additionally, CAISMOV24 cell line was characterized as a low-grade serous ovarian carcinoma, which still resembles its primary malignant cells.
Collapse
Affiliation(s)
| | | | - Gisele Olinto Libanio Rodrigues
- Instituto de Biologia, University of Campinas, Campinas, SP Brazil
- Laboratório de Biologia Molecular, Centro Infantil Boldrini, Campinas, SP Brazil
| | | | - Natacha Azussa Migita
- Instituto de Biologia, University of Campinas, Campinas, SP Brazil
- Laboratório de Biologia Molecular, Centro Infantil Boldrini, Campinas, SP Brazil
| | | | - Sophie Derchain
- Faculdade de Ciências Médicas, University of Campinas, Campinas, SP Brazil
- Women’s Hospital “Professor Doutor José Aristodemo Pinotti” – CAISM, University of Campinas, Rua Alexander Fleming 101, Campinas, SP 13083-881 Brazil
| | - José Andrés Yunes
- Faculdade de Ciências Médicas, University of Campinas, Campinas, SP Brazil
- Laboratório de Biologia Molecular, Centro Infantil Boldrini, Campinas, SP Brazil
| | - Fernando Guimarães
- Faculdade de Ciências Médicas, University of Campinas, Campinas, SP Brazil
- Women’s Hospital “Professor Doutor José Aristodemo Pinotti” – CAISM, University of Campinas, Rua Alexander Fleming 101, Campinas, SP 13083-881 Brazil
| |
Collapse
|
36
|
Sadlecki P, Antosik P, Grzanka D, Grabiec M, Walentowicz-Sadlecka M. KRAS mutation testing in borderline ovarian tumors and low-grade ovarian carcinomas with a rapid, fully integrated molecular diagnostic system. Tumour Biol 2017; 39:1010428317733984. [DOI: 10.1177/1010428317733984] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Pawel Sadlecki
- Department of Obstetrics and Gynecology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Paulina Antosik
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Marek Grabiec
- Department of Obstetrics and Gynecology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Malgorzata Walentowicz-Sadlecka
- Department of Obstetrics and Gynecology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| |
Collapse
|
37
|
Gwiti P, Vereczkey I, Cundell D, Aslam S, Clench T, Csernák E, Götzer K, Braybrooke J, Sohail M, Melegh Z. The mutational frequency of BRAF and KRAS in low-grade serous testicular neoplasms-a case series. Histopathology 2017; 71:686-692. [PMID: 28543997 DOI: 10.1111/his.13261] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/11/2017] [Accepted: 05/17/2017] [Indexed: 01/18/2023]
Abstract
AIMS Low-grade serous neoplasms of the testis are rare neoplasms that show striking morphological similarities with the better-understood ovarian neoplasms. This study is to see if there are similar molecular abnormalities in these two tumours. The cell of origin, relationship with serous ovarian tumour and the pathogenesis of these neoplasms are not fully established. METHODS AND RESULTS As low-grade serous ovarian neoplasms are known to harbour mutations in the MAPK pathway, we investigated the involvement of BRAF and KRAS mutations in low-grade testicular serous tumour by performing mutational analysis of seven cases. Mutational analysis was performed by melting curve analysis followed by bidirectional sequencing. Our findings showed BRAF and/or KRAS mutations in three of the seven cases, which is similar to the proportions reported in low-grade ovarian serous neoplasms. Of these three cases, one showed co-mutation of BRAF and KRAS. CONCLUSION The findings of this study are in support of a role of aberrant signalling of the MAPK pathway in the pathogenesis of low-grade serous testicular neoplasms, and provide a genetic link between low-grade testicular and ovarian serous tumours.
Collapse
Affiliation(s)
| | - Ildikó Vereczkey
- Department of Surgical and Molecular Pathology, National Institute of Oncology, Budapest, Hungary
| | - David Cundell
- Department of Pathology, Heart of England NHS Foundation Trust, Birmingham, UK
| | - Shazia Aslam
- Molecular Haematology Department, Bristol Royal Infirmary, Bristol, UK
| | - Tim Clench
- Molecular Haematology Department, Bristol Royal Infirmary, Bristol, UK
| | - Erzsébet Csernák
- Department of Surgical and Molecular Pathology, National Institute of Oncology, Budapest, Hungary
| | - Katalin Götzer
- Department of Surgical and Molecular Pathology, National Institute of Oncology, Budapest, Hungary
| | | | | | - Zsombor Melegh
- Department of Pathology, Southmead Hospital, Bristol, UK
| |
Collapse
|
38
|
Corassa M, Guimarães APG, Sanches SM, Fanelli MF, Rocha BMM, da Costa AABA, Alves V, Baiocchi G, Chinen LTD. Circulating tumor cells as a new and additional approach to follow-up patients with serous low-grade ovarian adenocarcinoma – a case report and review of the literature. ACTA ACUST UNITED AC 2017. [DOI: 10.1186/s41241-017-0030-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
39
|
18 F-FDG PET/CT as predictor of tumor biology and prognosis in epithelial ovarian carcinoma. Rev Esp Med Nucl Imagen Mol 2017. [DOI: 10.1016/j.remnie.2017.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Mert I, Chhina J, Allo G, Dai J, Seward S, Carey MS, Llaurado M, Giri S, Rattan R, Munkarah AR. Synergistic effect of MEK inhibitor and metformin combination in low grade serous ovarian cancer. Gynecol Oncol 2017; 146:319-326. [PMID: 28545687 DOI: 10.1016/j.ygyno.2017.05.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/09/2017] [Accepted: 05/13/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Low-grade serous ovarian cancer (LGSOC) constitutes 5-8% of epithelial ovarian cancers and is refractory to chemotherapy. We and others have shown metformin to cause significant growth inhibition in high-grade ovarian cancer both in vitro and in vivo. Here, we aimed to analyze if metformin was effective in inhibiting proliferation of LGSOC alone and in combination with MEK inhibitor. METHODS Three LGSOC lines (VOA1056, VOA1312 and VOA5646) were treated with metformin, trametinib or 2-deoxyglucose (2DG) alone or in combination with metformin. Proliferation was measured by MTT assay over a period of four days. Protein expression was measured by western blotting. Seahorse Analyzer was used to measure effect of metformin on glycolysis and mitochondrial respiration. RESULTS All LGSOC cell lines showed significant inhibition with metformin in a dose- and time-dependent manner. Trametinib significantly inhibited the growth of Ras mutated LGSOC lines (VOA1312 and VOA1056), while VOA5646 cells without RAS mutation did not show any response. Metformin and trametinib combination showed synergistic inhibition of RAS mutated VOA1312 and VOA1056 cells, but not for non-Ras mutated VOA5646 cells. Metformin and trametinib increased phosphorylated AMPK expression in LGSOC lines with combination showing stronger expression. Trametinib decreased 42/44 mitogen activated kinase phosphorylation in all cell lines, while metformin and combination had no significant effect. 2-DG significantly inhibited glycolysis in all LGSOC lines and combination with metformin showed synergistic inhibitory effect. CONCLUSIONS Metformin alone or in combination with MEK and glycolytic inhibitors may be a potential therapy for LGSOC, a cancer that is indolent but chemo-resistant.
Collapse
Affiliation(s)
- Ismail Mert
- Wayne State University, Department of Obstetrics and Gynecology, Detroit, MI, USA
| | - Jasdeep Chhina
- Henry Ford Health System, Division of Gynecologic Oncology, Detroit, MI, USA
| | - Ghassan Allo
- Henry Ford Health System, Department of Pathology, Detroit, MI, USA
| | - Jing Dai
- Wayne State University, Department of Obstetrics and Gynecology, Detroit, MI, USA
| | - Shelly Seward
- Wayne State University, Department of Obstetrics and Gynecology, Detroit, MI, USA
| | - Mark S Carey
- University of British Columbia, Vancouver, BC, Canada
| | | | - Shailendra Giri
- Henry Ford Health System, Department of Neurology, Detroit, MI, USA
| | - Ramandeep Rattan
- Henry Ford Health System, Division of Gynecologic Oncology, Detroit, MI, USA.
| | - Adnan R Munkarah
- Henry Ford Health System, Division of Gynecologic Oncology, Detroit, MI, USA.
| |
Collapse
|
41
|
Buttarelli M, Mascilini F, Zannoni GF, Ciucci A, Martinelli E, Filippetti F, Scambia G, Ferrandina G, Gallo D. Hormone receptor expression profile of low-grade serous ovarian cancers. Gynecol Oncol 2017; 145:352-360. [DOI: 10.1016/j.ygyno.2017.02.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/30/2022]
|
42
|
González García B, García Vicente AM, Jiménez Londoño GA, Pena Pardo FJ, Bellón Guardia ME, Talavera Rubio MP, Palomar Muñoz A, Gómez Herrero P, Soriano Castrejón ÁM. 18F-FDG PET/CT as predictor of tumour biology and prognosis in epithelial ovarian carcinoma. Rev Esp Med Nucl Imagen Mol 2017; 36:233-240. [PMID: 28284928 DOI: 10.1016/j.remn.2017.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/05/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To investigate the relationship between maximum standardised uptake value (SUVmax) of ovarian lesions and histopathology subtypes, and their involvement in the response and prognosis of patients with epithelial ovarian carcinoma (EOC). MATERIAL AND METHODS A retrospective analysis of 31 patients with EOC and 18F-FDG-PET/CT before treatment, including an assessment of the SUVmax of ovarian lesion. Histopathological diagnosis and follow-up was performed. A study was made on the relationship between the SUVmax and histological type (type I and II) and tumour stage, as well as the role of various parameters (SUVmax, histology, stage) on the patient outcomes (complete response [CR], overall survival [OS], disease-free survival [DFS], and disease-free [DF] status, at 12 and 24 months). RESULTS The medium SUVmax in type I lesions was lower than in type II (6.3 and 9.3, respectively; P=.03). A 7.1 cut-off was set for SUVmax in order to identify type II EOC (sensitivity: 77.8%, specificity: 69.2%; AUC=0.748; P=.02). No significant relationship was found between tumour stage and SUVmax. CR was more common in early stages; relative risk (RR) of 1.64; P=.003, as well as in type I tumours and a lower SUVmax. Tumour stage was decisive in DFS (P=.04), LE24m (0.07) and OS (P=.08). Longer DFS and a higher percentage of DF 24m were observed in type I tumours (RR: 1.32; P=.26). CONCLUSIONS SUVmax was related to EOC histology, so could predict the response and prognosis of these patients. No association was found between glycolytic activity of the primary tumor with the response and prognosis.
Collapse
Affiliation(s)
| | | | | | - F J Pena Pardo
- Hospital General Universitario Ciudad Real, Ciudad Real, España
| | | | | | - A Palomar Muñoz
- Hospital General Universitario Ciudad Real, Ciudad Real, España
| | - P Gómez Herrero
- Hospital General Universitario Ciudad Real, Ciudad Real, España
| | | |
Collapse
|
43
|
Recurrent low grade serous ovarian cancer in a 20 year old woman: A case from the Ohio State University College of Medicine. Gynecol Oncol 2017; 144:451-455. [DOI: 10.1016/j.ygyno.2017.01.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
44
|
Feng Z, Wen H, Ju X, Bi R, Chen X, Yang W, Wu X. Expression of hypothalamic-pituitary-gonadal axis-related hormone receptors in low-grade serous ovarian cancer (LGSC). J Ovarian Res 2017; 10:7. [PMID: 28122595 PMCID: PMC5264293 DOI: 10.1186/s13048-016-0300-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023] Open
Abstract
Background The aim of our study was to investigate the clinical features and expression levels of hypothalamic-pituitary-gonadal axis-related hormone receptors in low-grade serous ovarian cancer (LGSC). Methods We retrospectively investigated the clinical features of 26 consecutive patients with LGSC who underwent primary staging or debulking surgery between April 2005 and June 2013 in our center; concomitant primary high-grade serous ovarian cancer (HGSC) patients were randomly selected at a 2:1 ratio for comparison. Tissue microarrays were constructed from the LGSC and HGSC specimens, and the expression levels of six hormone receptors in the hypothalamic pituitary-gonadal axis were analyzed by immunohistochemistry. Results The median (range) age of patients with LGSC was 54 (27–77) years. According to the FIGO staging system, the cases were distributed as follows: stage I, 6 (23.1%); stage II, 0 (0%); stage III, 19 (73.1%); and stage IV, 1 (3.8%). The 2-year and 5-year overall survival rates for LGSC were 91.8% and 67.5%, respectively. The expression levels of the hormone receptors were as follows: ER, 80.8%; PR, 34.6%; AR, 53.8%; FSHR, 84.0%; LHR, 65.4%; and GnRHR, 100%. Hormone receptor-positive patients had a better prognosis compared with hormone receptor-negative patients, but the difference was not significant. Conclusions Our study presented a higher overall survival rate and distinctive hormone receptor expression levels of LGSC patients compared with the HGSC cohort. Patients with positive hormone receptor expression tended to have a better prognosis than the corresponding hormone receptor negative patients. Electronic supplementary material The online version of this article (doi:10.1186/s13048-016-0300-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zheng Feng
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China
| | - Hao Wen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China
| | - Xingzhu Ju
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rui Bi
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaojun Chen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wentao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaohua Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
45
|
Rossi ED, Martini M, Bizzarro T, Schmitt F, Longatto-Filho A, Larocca LM. Somatic mutations in solid tumors: a spectrum at the service of diagnostic armamentarium or an indecipherable puzzle? The morphological eyes looking for BRAF and somatic molecular detections on cyto-histological samples. Oncotarget 2017; 8:3746-3760. [PMID: 27738305 PMCID: PMC5356915 DOI: 10.18632/oncotarget.12564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 10/03/2016] [Indexed: 12/12/2022] Open
Abstract
This review article deals with the analysis and the detection of the morphological features associated with somatic mutations, mostly BRAFV600E mutation, on both cytological and histological samples of carcinomas. Few authors demonstrated that some architectural and specific cellular findings (i.e. polygonal eosinophilic cells defined as "plump cells" and sickle-shaped nuclei) are able to predict BRAF V600E mutation in both cytological and histological samples of papillary thyroid carcinoma (PTC) as well as in other carcinomas. In the current review article we evaluated the first comprehensive analysis of the morphological prediction of BRAFV600E and other somatic mutations in different malignant lesions with the description of the possible mechanisms beneath these morphologic features. The detection of predictive morphological features, mostly on FNAC, may add helpful information to the stratification of the malignant risk and personalized management of cancers. Additionally, the knowledge of the molecular mechanism of different oncogenic drivers can lead to the organ-specific triaging selection of cases and can provide significant insight for targeted therapies in different malignant lesions.
Collapse
Affiliation(s)
- Esther Diana Rossi
- Division of Anatomic Pathology and Histology, Università Cattolica del Sacro Cuore, “Agostino Gemelli” School of Medicine, Rome, Italy
| | - Maurizio Martini
- Division of Anatomic Pathology and Histology, Università Cattolica del Sacro Cuore, “Agostino Gemelli” School of Medicine, Rome, Italy
| | - Tommaso Bizzarro
- Division of Anatomic Pathology and Histology, Università Cattolica del Sacro Cuore, “Agostino Gemelli” School of Medicine, Rome, Italy
| | - Fernando Schmitt
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Department of Medicine and Pathology, Laboratoire National de Santé, Luxembourg
| | - Adhemar Longatto-Filho
- Department of Pathology, Laboratory of Medical Investigation, University of São Paulo School of Medicine, Brazil
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Pio XII Foundation, Barretos, Brazil
| | - Luigi Maria Larocca
- Division of Anatomic Pathology and Histology, Università Cattolica del Sacro Cuore, “Agostino Gemelli” School of Medicine, Rome, Italy
| |
Collapse
|
46
|
Rojas V, Hirshfield KM, Ganesan S, Rodriguez-Rodriguez L. Molecular Characterization of Epithelial Ovarian Cancer: Implications for Diagnosis and Treatment. Int J Mol Sci 2016; 17:E2113. [PMID: 27983698 PMCID: PMC5187913 DOI: 10.3390/ijms17122113] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 12/27/2022] Open
Abstract
Epithelial ovarian cancer is a highly heterogeneous disease characterized by multiple histological subtypes. Molecular diversity has been shown to occur within specific histological subtypes of epithelial ovarian cancer, between different tumors of an individual patient, as well as within individual tumors. Recent advances in the molecular characterization of epithelial ovarian cancer tumors have provided the basis for a simplified classification scheme in which these cancers are classified as either type I or type II tumors, and these two categories have implications regarding disease pathogenesis and prognosis. Molecular analyses, primarily based on next-generation sequencing, otherwise known as high-throughput sequencing, are allowing for further refinement of ovarian cancer classification, facilitating the elucidation of the site(s) of precursor lesions of high-grade serous ovarian cancer, and providing insight into the processes of clonal selection and evolution that may be associated with development of chemoresistance. Potential therapeutic targets have been identified from recent molecular profiling studies of these tumors, and the effectiveness and safety of a number of specific targeted therapies have been evaluated or are currently being studied for the treatment of women with this disease.
Collapse
Affiliation(s)
- Veronica Rojas
- Department Obstetrics/Gynecology and Reproductive Sciences, Rutgers Robert Wood Johnson Medical School, 125 Paterson Street, New Brunswick, NJ 08901, USA.
| | - Kim M Hirshfield
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
- Precision Medicine Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
| | - Shridar Ganesan
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
- Precision Medicine Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
| | - Lorna Rodriguez-Rodriguez
- Precision Medicine Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
- Department Obstetrics/Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
| |
Collapse
|
47
|
Tang W, Ren A, Xiao H, Sun H, Li B. Highly expressed NRSN2 is related to malignant phenotype in ovarian cancer. Biomed Pharmacother 2016; 85:248-255. [PMID: 27908706 DOI: 10.1016/j.biopha.2016.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/21/2016] [Accepted: 11/03/2016] [Indexed: 11/30/2022] Open
Abstract
Neurensin-2 (NRSN2) is a 24KD protein, which is reported located in the membrane, while its biological functions remain unknown, not to mention in the field of tumor biology. In current study, we aimed to analyze the functions of NRSN2 in ovarian cancer. We screened TCGA database and surprisingly found that its copy number and mRNA level are gained and heightened respectively in parts of serous ovarian cancer patients. In current study, both loss- and gain- function assays found that NRSN2 is associated with the malignant phenotype in ovarian cancer cells, because NRSN2 plays a remarkable role in anchorage-independent colony formation, subcutaneous tumor formation, cell invasion, and chemoresistance. Furthermore, we found that the level of NRSN2 was positively correlated with the expression of stem cell marker CD133. In addition, Wnt canonical signaling and Twist/Akt/Erk axis were also regulated by NRSN2. In conclusion, we found that a poorly studied protein, NRSN2, which is associated with the malignant phenotype of serous ovarian cancer and as a membrane protein; it could be a target for serous ovarian cancer treatment.
Collapse
Affiliation(s)
- Wenbin Tang
- The Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, China
| | - Aimin Ren
- The Department of Gynecology, Zhongshan Hospital Fudan University, China
| | - Hongyang Xiao
- The Department of Gynecology, Zhongshan Hospital Fudan University, China
| | - Huizhen Sun
- The Department of Gynecology, Zhongshan Hospital Fudan University, China
| | - Bin Li
- The Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, China.
| |
Collapse
|
48
|
Chen X, Wang X, Wei X, Wang J. EphA5 protein, a potential marker for distinguishing histological grade and prognosis in ovarian serous carcinoma. J Ovarian Res 2016; 9:83. [PMID: 27887627 PMCID: PMC5123222 DOI: 10.1186/s13048-016-0292-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/16/2016] [Indexed: 12/11/2022] Open
Abstract
Background Ovarian serous carcinoma (OSC) is the most common ovarian epithelial malignancy. Disregulation of Eph/ephrin signaling has been implicated in oncogenesis and tumor progression. EphA5 receptor is one of large families of Eph tyrosine kinase receptor and is documented in the development of nervous system. Till now, there is no published data about the role of EphA5 in ovarian epithelial neoplasmas. Methods This study aims to investigate the expression of EphA5 protein in ovarian serous carcinoma, and its relationship to clinical pathological characteristics. Sixty-one cases of ovarian serous carcinoma, 24 cases of benign ovarian serous tumors, 42 cases of serous borderline tumors and 20 cases of normal fallopian tubes were examined using immunohistochemical staining. The relationship between EphA5 expression and pathological parameters was analyzed. Kaplan-Meier survival function was used to analyze prognosis of patients. Results Immunostaining analysis demonstrated that the EphA5 protein was highly expressed in 100% (20/20) of normal fallopian tube samples, 100% (24/24) of benign epithelial ovarian tumors, 76% (32/42) of ovarian serous borderline tumors, and 31% (19/61) of ovarian serous carcinomas. Loss of EphA5expression was associated with tumor grade (P < 0.001) and FIGO stage (P = 0.005). The survival analysis showed that patients with negative or weak expression of EphA5 protein had a poor outcome than those with positive expression (P = 0.004). Conclusions Our results show that EphA5 may be a potential biomarker for distinguishing high-and low-grade ovarian serous carcinoma and a potential prognostic marker.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Xuan Wang
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Xue Wei
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Jiandong Wang
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China.
| |
Collapse
|
49
|
Ovarian cancer. Nat Rev Dis Primers 2016. [PMID: 27558151 DOI: 10.1038/nrdp.2016.61]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Ovarian cancer is not a single disease and can be subdivided into at least five different histological subtypes that have different identifiable risk factors, cells of origin, molecular compositions, clinical features and treatments. Ovarian cancer is a global problem, is typically diagnosed at a late stage and has no effective screening strategy. Standard treatments for newly diagnosed cancer consist of cytoreductive surgery and platinum-based chemotherapy. In recurrent cancer, chemotherapy, anti-angiogenic agents and poly(ADP-ribose) polymerase inhibitors are used, and immunological therapies are currently being tested. High-grade serous carcinoma (HGSC) is the most commonly diagnosed form of ovarian cancer and at diagnosis is typically very responsive to platinum-based chemotherapy. However, in addition to the other histologies, HGSCs frequently relapse and become increasingly resistant to chemotherapy. Consequently, understanding the mechanisms underlying platinum resistance and finding ways to overcome them are active areas of study in ovarian cancer. Substantial progress has been made in identifying genes that are associated with a high risk of ovarian cancer (such as BRCA1 and BRCA2), as well as a precursor lesion of HGSC called serous tubal intraepithelial carcinoma, which holds promise for identifying individuals at high risk of developing the disease and for developing prevention strategies.
Collapse
|
50
|
Matulonis UA, Sood AK, Fallowfield L, Howitt BE, Sehouli J, Karlan BY. Ovarian cancer. Nat Rev Dis Primers 2016. [PMID: 27558151 DOI: 10.1038/nrdp.2016.61] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Ovarian cancer is not a single disease and can be subdivided into at least five different histological subtypes that have different identifiable risk factors, cells of origin, molecular compositions, clinical features and treatments. Ovarian cancer is a global problem, is typically diagnosed at a late stage and has no effective screening strategy. Standard treatments for newly diagnosed cancer consist of cytoreductive surgery and platinum-based chemotherapy. In recurrent cancer, chemotherapy, anti-angiogenic agents and poly(ADP-ribose) polymerase inhibitors are used, and immunological therapies are currently being tested. High-grade serous carcinoma (HGSC) is the most commonly diagnosed form of ovarian cancer and at diagnosis is typically very responsive to platinum-based chemotherapy. However, in addition to the other histologies, HGSCs frequently relapse and become increasingly resistant to chemotherapy. Consequently, understanding the mechanisms underlying platinum resistance and finding ways to overcome them are active areas of study in ovarian cancer. Substantial progress has been made in identifying genes that are associated with a high risk of ovarian cancer (such as BRCA1 and BRCA2), as well as a precursor lesion of HGSC called serous tubal intraepithelial carcinoma, which holds promise for identifying individuals at high risk of developing the disease and for developing prevention strategies.
Collapse
Affiliation(s)
- Ursula A Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, and Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lesley Fallowfield
- Sussex Health Outcomes Research and Education in Cancer (SHORE-C), Brighton and Sussex Medical School, University of Sussex, Falmer, East Sussex, UK
| | - Brooke E Howitt
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jalid Sehouli
- Charité Universitaetsmedizin Berlin Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Beth Y Karlan
- Women's Cancer Program, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|