1
|
Benabdelkamel H, Jaber MA, Akkour K, AlMalki RH, Alfadda AA, Masood A, Joy SS, Alhalal H, Alwehaibi MA, Arafah M, Alshehri E, Abdel Rahman AM. Metabolomic Profiling of Blood Plasma in Females with Hyperplasia and Endometrial Cancer. Metabolites 2024; 14:109. [PMID: 38393001 PMCID: PMC10890097 DOI: 10.3390/metabo14020109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Uterine cancer is the most prevalent gynecologic malignancy in women worldwide. Endometrial cancer (EC) has an 81% five-year survival rate, depending on disease stage and time of diagnosis. While endometrial cancer is largely treatable when detected early, no established screening techniques are available in clinical practice. As a result, one of the most significant issues in the medical field is the development of novel ways for early cancer identification, which could boost treatment success rates. Liquid chromatography-high-resolution mass spectrometry (LC-HRMS)-based metabolomics was employed to explore the metabolomic markers and pathways unique to this cancer type and link them to the benign endometrial hyperplasia that may progress to cancer in 5% to 25% of patients. The study involved 59 postmenopausal participants, 20 with EC type 1, 20 with benign hyperplasia, and 19 healthy participants. Metabolite distribution changes were analyzed, and 338 of these features were dysregulated and significant. The first two main components, PC1 and PC2, were responsible for 11.5% and 12.2% of the total metabolites, respectively. Compared with the control group (CO), EC samples had 203 differentially expressed metabolites (180 upregulated and 23 downregulated); in hyperplasia (HP), 157 metabolites were dysregulated (127 upregulated and 30 downregulated) compared to the CO group while 21 metabolites exhibited differential regulation (16 upregulated and 5 downregulated) in EC plasma samples compared to the HP group. Hyperplasia samples exhibited similar metabolic changes to those reported in cancer, except for alterations in triglyceride levels, 7a,12 b-dihydroxy-5b-Cholan-24-oic acid, and Hept-2-enedioyl carnitine levels. The metabolites N-heptanoyl glycine and -(Methylthio)-2,3-isopentyl phosphate and formimino glutamic acid can be specific markers for hyperplasia conditions and dimethyl phosphatidyl ethanolamine and 8-isoprostaglandin E2 can be specific markers for EC conditions. Metabolic activities rely on mitochondrial oxidative phosphorylation for energy generation. The changes in metabolites identified in our study indicate that endometrial cancer cells adopt alternative strategies to increase energy production to meet the energy demand, thereby supporting proliferation.
Collapse
Affiliation(s)
- Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Malak A Jaber
- Pharmaceutical Medicinal Chemistry & Pharmacognosy, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 1196, Jordan
| | - Khalid Akkour
- Obstetrics and Gynecology Department, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Reem H AlMalki
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11461, Saudi Arabia
| | - Assim A Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Department of Medicine, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Salini Scaria Joy
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Hani Alhalal
- Obstetrics and Gynecology Department, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Moudi A Alwehaibi
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Maria Arafah
- Department of Pathology, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Eman Alshehri
- Obstetrics and Gynecology Department, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Anas M Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genome Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh 11211, Saudi Arabia
| |
Collapse
|
2
|
Exploration of the Molecular Mechanism of Danzhi Xiaoyao Powder in Endometrial Cancer through Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8330926. [PMID: 35774749 PMCID: PMC9239783 DOI: 10.1155/2022/8330926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/26/2022] [Indexed: 11/17/2022]
Abstract
Endometrial cancer (EC) is a common malignant tumor of the female reproductive system. Current treatments such as surgery and long-term hormone therapy are ineffective and have side effects. Danzhi Xiaoyao powder (DXP) can inhibit the growth of EC cells and induce apoptosis, but the pharmacological and molecular mechanisms of anticancer effects are still unclear. In this study, active components and potential targets of DXP were obtained from public databases. Protein effects and regulatory pathways of common targets were analyzed by protein-protein interaction (PPI), GO and KEGG. The results of network pharmacology showed that there are 87 common targets between EC and DXP. GO enrichment analysis showed that these targets were associated with response to oxidative stress, response to nutrient levels, hormone receptor binding and nuclear hormone receptor binding, etc. The results of KEGG analysis indicated that IL-17, TNF, PI3K/AKT, and RAS/RAF/MEK/ERK (ERK) signaling pathway were enriched in the anti-EC of DXP. Additionally, we cultured HEC-1B and KLE cells for validate experiments. DXP showed an inhibition of proliferation, migration, and cell cycle of both cells. Moreover, the expression of RAS, p-RAF, p-MEK, ERK, and p-ERK related proteins were downregulated. In conclusion, DXP might inhibit the proliferation of EC cells via apoptosis. Furthermore, DXP-induced inhibition of EC development might involve RAS/RAF/MEK/ERK pathway.
Collapse
|
3
|
Singla RK, Behzad S, Khan J, Tsagkaris C, Gautam RK, Goyal R, Chopra H, Shen B. Natural Kinase Inhibitors for the Treatment and Management of Endometrial/Uterine Cancer: Preclinical to Clinical Studies. Front Pharmacol 2022; 13:801733. [PMID: 35264951 PMCID: PMC8899191 DOI: 10.3389/fphar.2022.801733] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/17/2022] [Indexed: 02/05/2023] Open
Abstract
Endometrial cancer (EC) is the sixth most prevalent type of cancer among women. Kinases, enzymes mediating the transfer of adenosine triphosphate (ATP) in several signaling pathways, play a significant role in carcinogenesis and cancer cells’ survival and proliferation. Cyclin-dependent kinases (CDKs) are involved in EC pathogenesis; therefore, CDK inhibitors (CDKin) have a noteworthy therapeutic potential in this type of cancer, particularly in EC type 1. Natural compounds have been used for decades in the treatment of cancer serving as a source of anticancer bioactive molecules. Many phenolic and non-phenolic natural compounds covering flavonoids, stilbenoids, coumarins, biphenyl compounds, alkaloids, glycosides, terpenes, and terpenoids have shown moderate to high effectiveness against CDKin-mediated carcinogenic signaling pathways (PI3K, ERK1/2, Akt, ATM, mTOR, TP53). Pharmaceutical regimens based on two natural compounds, trabectedin and ixabepilone, have been investigated in humans showing short and midterm efficacy as second-line treatments in phase II clinical trials. The purpose of this review is twofold: the authors first provide an overview of the involvement of kinases and kinase inhibitors in the pathogenesis and treatment of EC and then discuss the existing evidence about natural products’ derived kinase inhibitors in the management of the disease and outline relevant future research.
Collapse
Affiliation(s)
- Rajeev K Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,IGlobal Research and Publishing Foundation, New Delhi, India
| | - Sahar Behzad
- Evidence-based Phytotherapy and Complementary Medicine Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Pharmacognosy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia.,Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
| | | | - Rupesh K Gautam
- Department of Pharmacology, MM School of Pharmacy, MM University, Ambala, India
| | - Rajat Goyal
- Department of Pharmacology, MM School of Pharmacy, MM University, Ambala, India
| | | | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Zeiter D, Vlajnic T, Schötzau A, Heinzelmann-Schwarz V, Montavon C. L1CAM is not a reliable predictor for lymph node metastases in endometrial cancer, but L1CAM positive patients benefit from radiotherapy. J Cancer 2021; 12:6401-6410. [PMID: 34659530 PMCID: PMC8489141 DOI: 10.7150/jca.59283] [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: 02/10/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose: Several studies evidenced the potential of L1CAM as a prognostic marker in endometrial cancer. The aim of this study was to investigate whether L1CAM can predict lymph node metastasis and could therefore be used preoperatively to identify patients with low to high-intermediate risk endometrial cancer who would profit from a lymphadenectomy and an adjuvant treatment. To avoid unnecessary morbidity, de-escalating strategies are still required. Methods: Immunohistochemistry for L1CAM was performed on curettage or hysterectomy specimens from 212 patients diagnosed with endometrial cancer who were treated at the University Hospital Basel during 2011-2019. L1CAM expression was correlated with clinicopathological features such as histological subtype, FIGO stage, lymph node metastasis, lymphadenectomy, adjuvant treatment and outcome. Results: Using a cut off ≥10%, L1CAM was positive in 41/212 patients (19.3%) and negative in 171/212 patients (80.7%). L1CAM was associated with high-risk features such as non-endometrioid histology, high tumour grade, and high FIGO stage. There was no significant correlation between L1CAM expression and lymph node metastasis. However, patients with L1CAM positive tumours showed improved disease-specific survival if treated with adjuvant radiotherapy. Conclusion: Although L1CAM expression pointed towards aggressive tumour biology, preoperative L1CAM analysis did not add any substantial predictive information regarding lymph node metastasis in low to high-intermediate risk groups. Therefore, L1CAM status is not suitable to tailor the surgical algorithm for lymph node staging. Nevertheless, our results suggest that L1CAM could be used as a predictive biomarker to select patients who may benefit the most from adjuvant radiotherapy.
Collapse
Affiliation(s)
- Deborah Zeiter
- Gynaecological Cancer Centre, Hospital for Women, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Tatjana Vlajnic
- Institute of Pathology, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Andreas Schötzau
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Viola Heinzelmann-Schwarz
- Gynaecological Cancer Centre, Hospital for Women, University Hospital Basel and University of Basel, 4031 Basel, Switzerland.,Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Céline Montavon
- Gynaecological Cancer Centre, Hospital for Women, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
5
|
Hernández JE, González-Montiel A, Allos-Villalva JCC, Cantú D, Barquet S, Olivares-Mundo A, Herrera LA, Prada D. Prognostic molecular biomarkers in endometrial cancer: A review. ACTA ACUST UNITED AC 2019; 7:17-28. [PMID: 34322276 PMCID: PMC8315102 DOI: 10.14312/2052-4994.2019-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background: Endometrial cancer (EC) is the fourth most common malignancy in women worldwide and the most common gynecological cancer in developed countries. The endometrioid subtype has an excellent prognosis with conventional treatment; however, recurrence reduces overall survival. Objective: Describe the most relevant evidence regarding selected potential molecular biomarkers that may predict overall survival (OS), relapse-free survival (RFS), and cancer-specific survival (CSS) in EC. Methods: An exhaustive search was performed in PUBMED with the search terms endometrial cancer, molecular biomarker, and survival. We selected original articles written in English about endometrial cancer, molecular biomarkers, and that included survival analysis published between January 2000 and December 2016. Results: Several molecular prognostic biomarkers have been studied in terms of survival and therapeutic response in women with endometrial cancer; hormone receptors, microRNAs, and other molecules have emerged as potentially useful biomarkers, including HER2, p21, HE4, PTEN, p27, ANCCA, and ANXA2. Conclusions: The use of biomarkers in the assessment of OS, RFS, and CSS requires large trials to expand our understanding of endometrial carcinogenesis. Several molecular markers are significantly associated with a high tumor grade and advanced clinical stage in EC and, therefore, could have additive effects when combined.
Collapse
Affiliation(s)
- J Edgardo Hernández
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Ailyn González-Montiel
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Jesús C Ceb Allos-Villalva
- Department of Biomedical Informatics, Faculty of Medicine, National Autonomous University of Mexico, C.U., Av. Universidad 3000, Mexico City, Mexico, 04510
| | - David Cantú
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Salim Barquet
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Anny Olivares-Mundo
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Luis A Herrera
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Diddier Prada
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080.,Department of Biomedical Informatics, Faculty of Medicine, National Autonomous University of Mexico, C.U., Av. Universidad 3000, Mexico City, Mexico, 04510
| |
Collapse
|
6
|
Anticancer Function and ROS-Mediated Multi-Targeting Anticancer Mechanisms of Copper (II) 2-hydroxy-1-naphthaldehyde Complexes. Molecules 2019; 24:molecules24142544. [PMID: 31336900 PMCID: PMC6680819 DOI: 10.3390/molecules24142544] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/06/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022] Open
Abstract
Multi-targeting of oncoproteins by a single molecule represents an effectual, rational, and an alternative approach to target therapy. We carried out a systematic study to reveal the mechanisms of action of newly synthesized Cu2+ compounds of 2-naphthalenol and 1-(((2-pyridinylmethyl)imino)methyl)- (C1 and C2). The antiproliferative activity of the as-synthesized complexes in three human cancer cell lines indicates their potential as multi-targeted antitumor agents. Relatively, C1 and C2 showed better efficacy in vitro relative to Cisplatin and presented promising levels of toxicity against A-549 cells. On the whole, the Cu2+ complexes exhibited chemotherapeutic effects by generating reactive oxygen species (ROS) and arresting the cell cycle in the G0/G1 phase by competent regulation of cyclin and cyclin-dependent kinases. Fascinatingly, the Cu2+ complexes were shown to activate the apoptotic and autophagic pathways in A-549 cells. These complexes effectively induced endoplasmic reticulum stress-mediated apoptosis, inhibited topoisomerase-1, and damaged cancer DNA through a ROS-mediated mechanism. The synthesized Cu2+ complexes established ROS-mediated targeting of multiple cell signaling pathways as a fabulous route for the inhibition of cancer cell growth.
Collapse
|
7
|
Ferlita AL, Battaglia R, Andronico F, Caruso S, Cianci A, Purrello M, Pietro CD. Non-Coding RNAs in Endometrial Physiopathology. Int J Mol Sci 2018; 19:ijms19072120. [PMID: 30037059 PMCID: PMC6073439 DOI: 10.3390/ijms19072120] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/12/2018] [Accepted: 07/14/2018] [Indexed: 12/18/2022] Open
Abstract
The Human Genome Project led to the discovery that about 80% of our DNA is transcribed in RNA molecules. Only 2% of the human genome is translated into proteins, the rest mostly produces molecules called non-coding RNAs, which are a heterogeneous class of RNAs involved in different steps of gene regulation. They have been classified, according to their length, into small non-coding RNAs and long non-coding RNAs, or to their function, into housekeeping non-coding RNAs and regulatory non-coding RNAs. Their involvement has been widely demonstrated in all cellular processes, as well as their dysregulation in human pathologies. In this review, we discuss the function of non-coding RNAs in endometrial physiology, analysing their involvement in embryo implantation. Moreover, we explore their role in endometrial pathologies such as endometrial cancer, endometriosis and chronic endometritis.
Collapse
Affiliation(s)
- Alessandro La Ferlita
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| | - Rosalia Battaglia
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| | - Francesca Andronico
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| | - Salvatore Caruso
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy.
| | - Antonio Cianci
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy.
| | - Michele Purrello
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| | - Cinzia Di Pietro
- Department of Biomedical and Biotechnological Sciences, Biology and Genetics Section G. Sichel, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
8
|
Kogata Y, Tanaka T, Ono YJ, Hayashi M, Terai Y, Ohmichi M. Foretinib (GSK1363089) induces p53-dependent apoptosis in endometrial cancer. Oncotarget 2018; 9:22769-22784. [PMID: 29854314 PMCID: PMC5978264 DOI: 10.18632/oncotarget.25232] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 04/06/2018] [Indexed: 12/17/2022] Open
Abstract
Objective Foretinib (GSK1363089 or XL880), which is an oral multikinase inhibitor developed to primarily target the hepatocyte growth factor (HGF)/Met signaling pathway, has shown anti-tumor effects against some cancers in preclinical and clinical studies. Results HGF/Met signaling in endometrial cancer cell lines was stimulated in an autocrine manner, and was essential for cell survival. Inhibiting the HGF/Met signaling with foretinib induced p53-dependent apoptosis in endometrial cancer cell lines in vitro. Foretinib also showed significant anti-cancer effects in vivo in experiments using cell tumor xenografts. p53 mutations were observed in 37 (10.8%) of 344 endometrial cancer specimens. Conclusion The HGF/Met-MAPK/PI3K pathway in endometrial cancer is activated by HGF in an autocrine manner. Foretinib induces an anti-cancer effect through the anti-phosphorylation of Met, which results in the induction of p53-dependent apoptosis; foretinib was found to exert greater anti-cancer activity in endometrial cancer specimens with wild-type p53 than in specimens with p53 mutations. Our immunochemical analysis revealed that foretinib-induced p53-dependent apoptosis can be expected to have therapeutic potential in approximately 90% of endometrial cancer patients. Methods We evaluated the HGF/Met signaling pathway in endometrial cancer cell lines and assessed the anti-cancer effects of foretinib using in vitro and in vivo experimental models. Furthermore, endometrial cancer specimens were subjected to an immunohistochemical analysis.
Collapse
Affiliation(s)
- Yuhei Kogata
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Japan
| | - Tomohito Tanaka
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Japan
| | - Yoshihiro J Ono
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Japan
| | - Masami Hayashi
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Japan
| | - Yoshito Terai
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Japan
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Osaka Medical College, Takatsuki, Japan
| |
Collapse
|
9
|
Non-Coding RNAs and Endometrial Cancer. Genes (Basel) 2018; 9:genes9040187. [PMID: 29596364 PMCID: PMC5924529 DOI: 10.3390/genes9040187] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 03/13/2018] [Accepted: 03/27/2018] [Indexed: 01/03/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are involved in the regulation of cell metabolism and neoplastic transformation. Recent studies have tried to clarify the significance of these information carriers in the genesis and progression of various cancers and their use as biomarkers for the disease; possible targets for the inhibition of growth and invasion by the neoplastic cells have been suggested. The significance of ncRNAs in lung cancer, bladder cancer, kidney cancer, and melanoma has been amply investigated with important results. Recently, the role of long non-coding RNAs (lncRNAs) has also been included in cancer studies. Studies on the relation between endometrial cancer (EC) and ncRNAs, such as small ncRNAs or micro RNAs (miRNAs), transfer RNAs (tRNAs), ribosomal RNAs (rRNAs), antisense RNAs (asRNAs), small nuclear RNAs (snRNAs), Piwi-interacting RNAs (piRNAs), small nucleolar RNAs (snoRNAs), competing endogenous RNAs (ceRNAs), lncRNAs, and long intergenic ncRNAs (lincRNAs) have been published. The recent literature produced in the last three years was extracted from PubMed by two independent readers, which was then selected for the possible relation between ncRNAs, oncogenesis in general, and EC in particular.
Collapse
|
10
|
Adjuvant chemoradiotherapy versus radiotherapy alone in high-risk endometrial cancer: A systematic review and meta-analysis. Gynecol Oncol 2018. [PMID: 29530332 DOI: 10.1016/j.ygyno.2018.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The benefits of adjuvant chemoradiotherapy (CRT) for high-risk endometrial cancer (HREC) in International Federation of Gynecology and Obstetrics (FIGO) stages I-III remain controversial. A systematic review and meta-analysis was conducted to evaluate the clinical effectiveness and safety of postoperative CRT over radiotherapy (RT) alone, exclusively for patients with HREC for the following key endpoints: overall survival (OS), progression-free survival (PFS), the local recurrence rate, the distant metastasis rate, cancer-specific survival (CSS), grade III/IV acute and late toxicities, and the small bowel obstruction rate. METHODS Five databases, namely, PubMed, EMBASE, Cochrane Library, Web of Science and ClinicalTrials.gov, were systematically explored and supplemented by manual searching to identify relevant studies published before Dec 9, 2017. Only prospective randomized controlled trials (RCTs) conducted for HREC comparing CRT and RT alone after surgery were included. All statistical analyses were performed using RevMan Version 5.3 software. RESULTS Six eligible trials involving 2105 patients were identified for the final meta-analysis (CRT: n = 1064; RT: n = 1041). No statistically significant differences were evident between the CRT and RT groups regarding OS (n = 2105, RR = 1.02, 95% CI 0.98-1.06, P = 0.40). Additionally, no differences were apparent in terms of the local recurrence rate (n = 690, RR = 0.48, 95% CI 0.19-1.18, P = 0.11) or distant metastasis rate (n = 1445, RR = 0.94, 95% CI 0.72-1.23, P = 0.67). However, CRT significantly prolonged overall five-year PFS (80.2% vs. 74.5%, +5.7%; RR = 1.08, P = 0.005) and five-year CSS (86.1% vs. 79.0%, +7.1%; RR = 1.09, P = 0.03). A higher incidence of grade III/IV toxicities (P < 0.00001) was evident with CRT, while grade III/IV late toxicities and the small bowel obstruction rate were not significantly different between the two groups. CONCLUSIONS For patients with endometrial cancers with stage I-III risk factors, adjuvant CRT can significantly improve PFS and CSS compared with RT. With the exception of increased acute toxicities, CRT is well accepted and tolerated in HREC patients.
Collapse
|
11
|
Survival outcomes and patterns of failure in women with stage IIIC2 endometrial carcinoma. Eur J Obstet Gynecol Reprod Biol 2017; 216:192-197. [DOI: 10.1016/j.ejogrb.2017.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 10/19/2022]
|
12
|
Sato E, Nakayama K, Nakamura K, Ishibashi T, Katagiri H, Ishikawa M, Kyo S. Bevacizumab plus chemotherapy continued beyond progression in patients with type II endometrial cancer previously treated with bevacizumab plus chemotherapy: A case report. Mol Clin Oncol 2017; 7:391-394. [PMID: 28781816 DOI: 10.3892/mco.2017.1316] [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: 09/14/2015] [Accepted: 02/28/2017] [Indexed: 01/06/2023] Open
Abstract
The prognosis of patients with recurrent/persistent endometrial cancer, particularly type II cancer, remains poor, and effective treatment has not yet been established. We herein present the case of a patient with recurrent type II endometrial cancer who received bevacizumab + chemotherapy continued beyond progression, after previously receiving bevacizumab + chemotherapy. This patient experienced recurrence after first- and second-line adjuvant chemotherapy followed by modified radical hysterectomy and she was administered bevacizumab + paclitaxel + carboplatin therapy. After six cycles of treatment, all metastatic lesions shrunk, indicating partial response. The patient next received single-agent bevacizumab as maintenance therapy. After 12 cycles of bevacizumab monotherapy, disease progression was detected; therefore, combination therapy consisting of bevacizumab, doxorubicin and carboplatin was initiated. After six cycles of this combination therapy, the patient exhibited disease stabilization. Finally, 18 months after the initial bevacizumab treatment, the patient remained on combination chemotherapy, without complaints or signs of tumor progression (last follow-up, October 2014).
Collapse
Affiliation(s)
- Emi Sato
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Kentaro Nakayama
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Kohei Nakamura
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Tomoka Ishibashi
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Hiroshi Katagiri
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Masako Ishikawa
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| |
Collapse
|
13
|
Winterhoff B, Konecny GE. Targeting fibroblast growth factor pathways in endometrial cancer. Curr Probl Cancer 2017; 41:37-47. [DOI: 10.1016/j.currproblcancer.2016.11.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
14
|
Nishimura T, Nakamura K, Yamashita S, Ikeda S, Kigure K, Minegishi T. Effect of the molecular targeted drug, erlotinib, against endometrial cancer expressing high levels of epidermal growth factor receptor. BMC Cancer 2015; 15:957. [PMID: 26673416 PMCID: PMC4682234 DOI: 10.1186/s12885-015-1975-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/05/2015] [Indexed: 11/29/2022] Open
Abstract
Background The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, erlotinib, has been clinically applied for the treatment of a variety of tumors with EGFR overexpression. A phase II clinical study of erlotinib (NCIC IND-148) for recurrent or metastatic endometrial carcinoma (EC) resulted in an unfavorable result. However, in that study, the expression levels of EGFR were not accurately analyzed. Thus, the aim of this study was to re-examine the efficacy of erlotinib in EC cells by utilizing in vitro and in vivo models. Methods Tissue samples obtained from patients histologically diagnosed with EC of the uterine corpus were subjected to immunohistochemistry and RT-PCR to determine the protein and mRNA expression levels of EGFR. Western blot and WST-1 assays of EGFR siRNA-transfected HEC-1A, KLE, and Ishikawa cells were used to evaluate the efficacy of erlotinib in tumor cell lines expressing different EGFR levels. Furthermore, HEC-1A and Ishikawa cells were implanted into athymic mice treated with either erlotinib or trastuzumab. Results At our institution, 20.9 % of endometrial cancer patients with low grade endometrioid histology have been diagnosed as stage III and IV. Immunohistochemical analysis and RT-PCR revealed the presence of significant EGFR and EGFR mRNA expression in low-grade endometrioid carcinoma in comparison with high-grade endometrioid carcinoma. In vitro study, WST-1 assay and Western blot analysis revealed that EGFR expression levels were correlated with tumor cell viability. Erlotinib reduced the proliferation of HEC-1A expressing high levels of EGFR, while trastuzumab showed similar effect in Ishikawa cells dominantly expressing human epidermal growth factor receptor type2 (HER2). In vivo erlotinib decreased tumor growth in mice xenografted with HEC-1A cells, whereas this tumor-growth inhibition was not observed in trastuzumab-treated mice xenografted with Ishikawa cell. Conclusions EGF contributed to tumor proliferation in EC cell lines along with EGFR expression in vitro. Erlotinib also demonstrated anti-tumor effects in xenograft mice models. Our results suggest that erlotinib continues to have clinical usefulness in specific cases, after taking into consideration the EGFR expression levels.
Collapse
Affiliation(s)
- Toshio Nishimura
- Department of Obstetrics and Gynecology, Gunma University, 3-39-22, Showa, Maebashi, Gunma, 371-8511, Japan.
| | - Kazuto Nakamura
- Gunma Prefectural Cancer Center, 617-1, Nishimachi, Takabayashi, Ota, Gunma, 373-8500, Japan.
| | - Soichi Yamashita
- Department of Obstetrics and Gynecology, Gunma University, 3-39-22, Showa, Maebashi, Gunma, 371-8511, Japan.
| | - Sadatomo Ikeda
- Department of Obstetrics and Gynecology, Gunma University, 3-39-22, Showa, Maebashi, Gunma, 371-8511, Japan.
| | - Keiko Kigure
- Gunma Prefectural Cancer Center, 617-1, Nishimachi, Takabayashi, Ota, Gunma, 373-8500, Japan.
| | - Takashi Minegishi
- Department of Obstetrics and Gynecology, Gunma University, 3-39-22, Showa, Maebashi, Gunma, 371-8511, Japan.
| |
Collapse
|
15
|
Chandra V, Kim JJ, Benbrook DM, Dwivedi A, Rai R. Therapeutic options for management of endometrial hyperplasia. J Gynecol Oncol 2015; 27:e8. [PMID: 26463434 PMCID: PMC4695458 DOI: 10.3802/jgo.2016.27.e8] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 07/24/2015] [Accepted: 07/31/2015] [Indexed: 12/24/2022] Open
Abstract
Endometrial hyperplasia (EH) comprises a spectrum of changes in the endometrium ranging from a slightly disordered pattern that exaggerates the alterations seen in the late proliferative phase of the menstrual cycle to irregular, hyperchromatic lesions that are similar to endometrioid adenocarcinoma. Generally, EH is caused by continuous exposure of estrogen unopposed by progesterone, polycystic ovary syndrome, tamoxifen, or hormone replacement therapy. Since it can progress, or often occur coincidentally with endometrial carcinoma, EH is of clinical importance, and the reversion of hyperplasia to normal endometrium represents the key conservative treatment for prevention of the development of adenocarcinoma. Presently, cyclic progestin or hysterectomy constitutes the major treatment option for EH without or with atypia, respectively. However, clinical trials of hormonal therapies and definitive standard treatments remain to be established for the management of EH. Moreover, therapeutic options for EH patients who wish to preserve fertility are challenging and require nonsurgical management. Therefore, future studies should focus on evaluation of new treatment strategies and novel compounds that could simultaneously target pathways involved in the pathogenesis of estradiol-induced EH. Novel therapeutic agents precisely targeting the inhibition of estrogen receptor, growth factor receptors, and signal transduction pathways are likely to constitute an optimal approach for treatment of EH.
Collapse
Affiliation(s)
- Vishal Chandra
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Jong Joo Kim
- School of Biotechnology, Yeungnam University, Gyeongsan, Korea
| | - Doris Mangiaracina Benbrook
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anila Dwivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajani Rai
- School of Biotechnology, Yeungnam University, Gyeongsan, Korea.
| |
Collapse
|
16
|
Long Non-Coding RNAs in Endometrial Carcinoma. Int J Mol Sci 2015; 16:26463-72. [PMID: 26556343 PMCID: PMC4661821 DOI: 10.3390/ijms161125962] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/22/2015] [Accepted: 10/26/2015] [Indexed: 12/01/2022] Open
Abstract
Endometrial carcinoma (EC), the second most common form of gynaecological malignancy, can be divided into two distinct sub-types: Type I tumours arise from hyperplastic endometrium and typically effect women around the time of menopause, whereas type II tumours arise in postmenopausal women from atrophic endometrium. Long non-coding RNAs (lncRNAs) are a novel class of non-protein coding molecules that have recently been implicated in the pathogenesis of many types of cancer including gynaecological tumours. Although they play critical physiological roles in cellular metabolism, their expression and function are deregulated in EC compared with paired normal tissue, indicating that they may also participate in tumour initiation and progression. For instance, the lncRNA MALAT-1 is down-regulated in EC samples compared to normal or hyperplastic endometrium, whereas the lncRNA OVAL is down-regulated in type II disease but up-regulated in type I disease. Other notatble lncRNAs such as HOTAIR, H19 and SRA become up-regulated with increasing EC tumour grade and other features associated with poor prognosis. In the current review, we will examine the growing body of evidence linking deregulated lncRNAs with specific biological functions of tumour cells in EC, we will highlight associations between lncRNAs and the molecular pathways implicated in EC tumourigenesis and we will identify critical knowledge gaps that remain to be addressed.
Collapse
|
17
|
Amant F, Mirza MR, Koskas M, Creutzberg CL. Cancer of the corpus uteri. Int J Gynaecol Obstet 2015; 131 Suppl 2:S96-104. [DOI: 10.1016/j.ijgo.2015.06.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
18
|
Seoud M, Lundqvist EÅ, Fujiwara K. Targeted therapy in gynecologic cancers: Ready for prime time? Int J Gynaecol Obstet 2015; 131 Suppl 2:S150-2. [DOI: 10.1016/j.ijgo.2015.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
19
|
Bogliolo S, Cassani C, Gardella B, Musacchi V, Babilonti L, Venturini PL, Ferrero S, Spinillo A. Current opinion on bevacizumab on endometrial cancer treatment. Expert Opin Biol Ther 2014; 15:299-307. [PMID: 25539414 DOI: 10.1517/14712598.2015.995624] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Advanced or recurrent endometrial cancer is still a challenge for clinicians as it has a poor prognosis despite treatment efforts. Thus, there is an urgent need for new agents with activity in this subset of patients. The increased knowledge of the molecular aspects of endometrial carcinogenesis has led to the development of molecular targeted therapies and in particular anti-angiogenic drugs. One of the most promising of these agents is bevacizumab , a recombinant humanized immunoglobulin monoclonal antibody to VEGF. AREAS COVERED The objective of this paper is to discuss the role of angiogenesis in endometrial cancer and analyze the rational of bevacizumab use, alone or in combination with other therapies, in endometrial cancer patients. We reviewed the most important preclinical and clinical studies published on this topic up to March 2014. EXPERT OPINION Bevacizumab in combination with others targeted therapies, chemotherapy or radiotherapy demonstrated promising anti-tumor activity. Despite the good oncological outcomes of these recent clinical experiences, caution must be used in light of significant toxicity reported in this subset of heavily pre-treated patients. The identification of biomarkers able to predict either the efficacy or toxicity of anti-angiogenic drugs is a compelling need.
Collapse
Affiliation(s)
- Stefano Bogliolo
- IRCCS-Fondazione Policlinico San Matteo and University of Pavia, Department of Obstetrics and Gynaecology , 19 Viale Camillo Golgi, 27100 Pavia , Italy
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Amadio G, Masciullo V, Ferrandina MG, Scambia G. Emerging drugs for endometrial cancer. Expert Opin Emerg Drugs 2014; 19:497-509. [PMID: 25330855 DOI: 10.1517/14728214.2014.971752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION From the dualistic classification that divides endometrial cancer (EC) into two types with distinct underlying molecular profiling, histopathology and clinical behavior, arises a deeper understanding of the carcinogenesis pathways. EC treatment comprises different and multimodal therapeutic approaches, such as chemotherapy, radiation therapy or combinations of novel drugs; however, few of these regimens have truly improved progression-free or survival rates in advanced and metastatic settings. AREAS COVERED We reviewed the main molecular pathways involved in EC carcinogenesis through a wide literature search of novel compounds that alone or in combination with traditional drugs have been investigated or are currently under investigation in randomized clinical trials. EXPERT OPINION The molecular therapies mainly discussed in this review are potential therapeutic candidates for more effective and specific treatments. In the genomic era, a deeper knowledge about molecular characteristics of cancer provides the hope for the development of better therapeutic approaches. Targeting both genetic and epigenetic alterations, attacking tumor cells using cell-surface markers overexpressed in tumor tissue, reactivating antitumor immune responses and identifying predictive biomarkers represent the emerging strategies and the major challenges.
Collapse
Affiliation(s)
- Giulia Amadio
- Catholic University of the Sacred Heart, Division of Gynecologic Oncology, Department of Gynecology and Obstetrics , Largo Agostino Gemelli 8, IT-00168 Rome , Italy +39 0630154979 ; +39 063051160 ;
| | | | | | | |
Collapse
|
21
|
Growth of poorly differentiated endometrial carcinoma is inhibited by combined action of medroxyprogesterone acetate and the Ras inhibitor Salirasib. Oncotarget 2014; 4:316-28. [PMID: 23530112 PMCID: PMC3712577 DOI: 10.18632/oncotarget.867] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Type 2 endometrial carcinoma (EC) is a poorly differentiated EC. Unlike type 1 EC, which responds to hormonal treatment (progestins), type 2 EC is refractory to hormonal treatment because of its low expression of active estrogen and progesterone receptors (ER, PR). The aim of this study was to develop a novel drug combination designed to treat these aggressive type 2 EC tumors without surgery and with fertility potential preserved. We examined the effects of combined treatment with the progestin medroxyprogesterone acetate (MPA) and the Ras inhibitor S-farnesylthiosalicylic acid (FTS; Salirasib). Because FTS can induce cell differentiation in tumor cells, we examined whether FTS could induce re-differentiation of type 2 EC cells, thereby sensitizing them to MPA. We found that FTS reduced Ras-GTP, phospho-Akt, and phospho-ERK, and that these reductions all correlated with a decrease in ERα phosphorylation. Combined treatment with FTS and MPA induced stronger reduction in USPC1 type 2 EC cell numbers than the reduction induced by either drug alone. MPA caused ERα degradation. Death of the cells was caused by MPA but not by FTS. The phosphorylated ERα induces gene transcription manifested by enhanced cell proliferation and survival. The combination of FTS and MPA, by reducing the mRNA expression of ERα-mediated genes (i.e. PR, c-fos and ps2/TFF1), inhibited tumor growth and enhanced the death of type 2 EC cells. These promising results might herald a novel treatment for the highly aggressive, incurable type 2 endometrial carcinoma.
Collapse
|
22
|
Lee PS, Secord AA. Targeting molecular pathways in endometrial cancer: a focus on the FGFR pathway. Cancer Treat Rev 2013; 40:507-12. [PMID: 24332498 DOI: 10.1016/j.ctrv.2013.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/08/2013] [Accepted: 11/12/2013] [Indexed: 01/18/2023]
Abstract
In the majority of cases, endometrial cancer is localized and highly curable through surgery and adjuvant radiotherapy. However, for patients with advanced or metastatic disease, prognosis is poor. Systemic treatments such as cytotoxic chemotherapy or hormonal therapy can cause significant toxicities including chemotherapy-related gastrointestinal, neurologic, and immunosuppressive toxicities and hormone-related hypertension, increased blood sugar, thrombosis, and pulmonary emboli. In addition, these therapies rarely lead to sustained disease control. Novel therapies with greater efficacy and reduced toxicity are needed. Recent progress in the identification of genetic abnormalities in cell signaling proteins has spurred the development of targeted agents for the treatment of patients with endometrial cancer. The fibroblast growth factor receptor (FGFR) pathway is one of several signaling pathways that have been implicated in the pathogenesis and progression of endometrial cancer. The activity of novel FGFR-targeted agents in preclinical endometrial cancer models and clinical trials will be reviewed.
Collapse
Affiliation(s)
- Paula S Lee
- Duke University Medical Center (DUMC) 3079, Durham, NC 27710, United States.
| | | |
Collapse
|
23
|
Subramaniam KS, Tham ST, Mohamed Z, Woo YL, Mat Adenan NA, Chung I. Cancer-associated fibroblasts promote proliferation of endometrial cancer cells. PLoS One 2013; 8:e68923. [PMID: 23922669 PMCID: PMC3724864 DOI: 10.1371/journal.pone.0068923] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/03/2013] [Indexed: 01/25/2023] Open
Abstract
Endometrial cancer is the most commonly diagnosed gynecologic malignancy worldwide; yet the tumor microenvironment, especially the fibroblast cells surrounding the cancer cells, is poorly understood. We established four primary cultures of fibroblasts from human endometrial cancer tissues (cancer-associated fibroblasts, CAFs) using antibody-conjugated magnetic bead isolation. These relatively homogenous fibroblast cultures expressed fibroblast markers (CD90, vimentin and alpha-smooth muscle actin) and hormonal (estrogen and progesterone) receptors. Conditioned media collected from CAFs induced a dose-dependent proliferation of both primary cultures and cell lines of endometrial cancer in vitro (175%) when compared to non-treated cells, in contrast to those from normal endometrial fibroblast cell line (51%) (P<0.0001). These effects were not observed in fibroblast culture derived from benign endometrial hyperplasia tissues, indicating the specificity of CAFs in affecting endometrial cancer cell proliferation. To determine the mechanism underlying the differential fibroblast effects, we compared the activation of PI3K/Akt and MAPK/Erk pathways in endometrial cancer cells following treatment with normal fibroblasts- and CAFs-conditioned media. Western blot analysis showed that the expression of both phosphorylated forms of Akt and Erk were significantly down-regulated in normal fibroblasts-treated cells, but were up-regulated/maintained in CAFs-treated cells. Treatment with specific inhibitors LY294002 and U0126 reversed the CAFs-mediated cell proliferation (P<0.0001), suggesting for a role of these pathways in modulating endometrial cancer cell proliferation. Rapamycin, which targets a downstream molecule in PI3K pathway (mTOR), also suppressed CAFs-induced cell proliferation by inducing apoptosis. Cytokine profiling analysis revealed that CAFs secrete higher levels of macrophage chemoattractant protein (MCP)-1, interleukin (IL)-6, IL-8, RANTES and vascular endothelial growth factor (VEGF) than normal fibroblasts. Our data suggests that in contrast to normal fibroblasts, CAFs may exhibit a pro-tumorigenic effect in the progression of endometrial cancer, and PI3K/Akt and MAPK/Erk signaling may represent critical regulators in how endometrial cancer cells respond to their microenvironment.
Collapse
Affiliation(s)
- Kavita S. Subramaniam
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Seng Tian Tham
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Pharmacogenomics Laboratory, Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yin Ling Woo
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Azmi Mat Adenan
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail:
| |
Collapse
|
24
|
Thanapprapasr D, Cheewakriangkrai C, Likittanasombut P, Thanapprapasr K, Mutch DG. Targeted endometrial cancer therapy as a future prospect. ACTA ACUST UNITED AC 2013; 9:189-99. [PMID: 23477324 DOI: 10.2217/whe.13.4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Among female-specific cancers worldwide, endometrial cancer is the third most common after breast cancer and cervical cancer. In addition, it is the most common gynecological cancer in the USA and Europe. The incidence of this disease appears to be increasing. The cause of this increase is multifactorial, but a few possible factors involved are increasing obesity, an aging population leading to more postmenopausal women and greater tamoxifen use. Surgery is generally the primary treatment of this disease and postoperative radiation therapy in some patients with high or intermediate risk may prevent locoregional recurrences. Adjuvant chemotherapy improves progression-free survival in advanced or recurrent cancer. However, overall survival in patients with advanced disease is poor. Hence, better therapy is needed and targeted molecular therapies are emerging as possible treatment candidates. These include molecules that target VEGF, mTOR, tyrosine kinases, human EGF receptors and FGF receptors. Therapies targeting specific molecular features should be evaluated in future strategies in the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Duangmani Thanapprapasr
- Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | | | | | | | | |
Collapse
|
25
|
The benefit of adjuvant chemotherapy combined with postoperative radiotherapy for endometrial cancer: a meta-analysis. Eur J Obstet Gynecol Reprod Biol 2013; 170:39-44. [PMID: 23810000 DOI: 10.1016/j.ejogrb.2013.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 03/31/2013] [Accepted: 06/05/2013] [Indexed: 11/20/2022]
Abstract
The objective of our study was to determine whether adjuvant chemotherapy combined with postoperative radiotherapy would have benefits for the disease-free survival and overall survival in patients with high-risk endometrial cancer. Electronic searches for studies of adjuvant chemotherapy combined with postoperative radiotherapy in endometrial cancer patients between March 1971 and March 2012 were made on MEDLINE, SCOPUS, and the Cochrane library. Articles with more than 4 stars on the Newcastle-Ottawa scale or a score of more than 4 on the modified Jadad scale were included. A meta-analysis was performed, and pooled hazard ratios (HR) of progression-free survival (PFS) and overall survival (OS) between patients whose adjuvant chemotherapy was combined with radiotherapy (the CTx+RTx group) and patients with adjuvant radiotherapy only (the RTx group) were derived from the fixed effect model or random effect model. Three observational studies and 3 randomized clinical trials (RCTs) were included in the final analysis. Subgroup analysis for FIGO stage showed that the CTx+RTx group had a more significant survival benefit compared to that of the RTx group in advanced stage endometrial cancer (OS HR 0.53, 95% CI 0.36-0.80; PFS HR 0.54, 95% CI 0.37-0.77), but no significant benefit in early stage endometrial cancer (OS HR 0.96, 95% CI 0.70-1.32; PFS HR 1.00, 95% CI 0.39-2.58). This meta-analysis suggests that adjuvant chemotherapy combined with postoperative radiotherapy could probably reduce disease progression and overall death in patients with advanced-stage disease. In order to examine whether the multimodal treatment has benefit in high-risk endometrial cancer, we need further large-scale RCTs.
Collapse
|
26
|
Nogami Y, Banno K, Kisu I, Yanokura M, Umene K, Masuda K, Kobayashi Y, Yamagami W, Nomura H, Tominaga E, Susumu N, Aoki D. Current status of molecular-targeted drugs for endometrial cancer (Review). Mol Clin Oncol 2013; 1:799-804. [PMID: 24649249 PMCID: PMC3915661 DOI: 10.3892/mco.2013.140] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 06/07/2013] [Indexed: 11/15/2022] Open
Abstract
Endometrial cancer is a common gynecological malignant tumor in Western countries and its incidence has also been on the increase in Asia. Genetic abnormalities related to onset and progression of malignancy in the endometrial membrane and signaling system have been identified and the developmental mechanism of endometrial cancer is becoming elucidated. The identification of the molecules related to these abnormalities has led to new potential treatment regimens for endometrial cancer, using molecular-targeted drugs. The current chemotherapy for endometrial cancer often causes systemic side effects that require discontinuation of the treatment. Furthermore, a treatment regimen for cancers of rare histological types has not been established. Recent studies on endometrial cancer revealed patterns of genetic disorders that differ among the histological types. Genetic and molecular information that underlie pathological changes and is associated with DNA mismatch repair genes and epigenetic regulation was also identified. Targeting of these mechanisms with molecular-targeted drugs has been performed with the aim of linking treatment to the carcinogenic mechanism at the molecular and genetic levels. However, the response rates with single-agent therapy are generally low and several problems remain unresolved. Trials of combinations of molecular-targeted drugs with currently available treatments and identification of factors determining sensitivity are required to overcome these difficulties.
Collapse
Affiliation(s)
- Yuya Nogami
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Iori Kisu
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Megumi Yanokura
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Kiyoko Umene
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Kenta Masuda
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Yusuke Kobayashi
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Eiichiro Tominaga
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Nobuyuki Susumu
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 1608582, Japan
| |
Collapse
|
27
|
|
28
|
Leslie KK, Thiel KW, Goodheart MJ, De Geest K, Jia Y, Yang S. Endometrial cancer. Obstet Gynecol Clin North Am 2012; 39:255-68. [PMID: 22640714 DOI: 10.1016/j.ogc.2012.04.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Despite the questions and barriers, the incorporation of molecular therapy into treatment regimens in endometrial cancer is an exciting area of investigation with the potential to improve outcomes. Outside of the development of a reliable screening test for endometrial cancer, converting the disease to a chronic state and improving progression-free survival is our best hope to reverse the concerning trend of decreasing 5-year survival for this disease.
Collapse
Affiliation(s)
- Kimberly K Leslie
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Gynecologic malignancies carry an estimated incidence of 83,750 cases per year and estimated mortality rate of more than 27,000 women per year. New therapies and therapeutic approaches are needed to improve the outlook for women with gynecologic cancers. Recent insights at the molecular and cellular levels are paving the way for a more directed approach to target mechanisms driving tumorigenesis. This article reviews the roles of new and emerging antiangiogenesis drugs, summarizes the data obtained from clinical trials of antiangiogenic agents, and discusses trials under way to address the role of such strategies in gynecologic cancers.
Collapse
Affiliation(s)
- Behrouz Zand
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030, USA
| | | | | |
Collapse
|
30
|
Abstract
BACKGROUND This is an updated version of the original Cochrane review published in Issue 2, 2007. The role of radiotherapy (both pelvic external beam radiotherapy (EBRT) and vaginal intracavity brachytherapy (VBT)) in stage I endometrial cancer following hysterectomy remains controversial. OBJECTIVES To assess the efficacy of adjuvant radiotherapy following surgery for stage I endometrial cancer. SEARCH METHODS We searched The Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE and the Specialised Register to end-2005 for the original review, and extended the search to January 2012 for the update. SELECTION CRITERIA We included randomised controlled trials (RCTs) that compared post-operative adjuvant radiotherapy (either EBRTor VBT, or both) versus no radiotherapy or VBT in women with stage I endometrial cancer. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trials and extracted data to a specifically designed data collection form. The primary outcome was overall survival. Secondary outcomes were endometrial cancer-related deaths, locoregional recurrence and distant recurrence. Meta-analyses were performed using Cochrane Review Manager Software 5.1. MAIN RESULTS We included eight trials. Seven trials (3628 women) compared EBRT with no EBRT (or VBT), and one trial (645 women) compared VBTwith no additional treatment. We considered six of the eight trials to be of a high quality. Time-to-event data were not available for all trials and all outcomes.EBRT (with or without VBT) compared with no EBRT (or VBT alone) for stage I endometrial carcinoma significantly reduced locoregional recurrence (time-to-event data: five trials, 2965 women; Hazard Ratio (HR) 0.36, 95% Confidence Interval (CI) 0.25 to 0.52; and dichotomous data: seven trials, 3628 women; Risk Ratio (RR) 0.33, 95% CI 0.23 to 0.47). This reduced risk of locoregional recurrence did not translate into improved overall survival (time-to-event data: five trials, 2,965 women; HR 0.99, 95% CI 0.82 to1.20; and dichotomous data: seven trials, 3628 women; RR 0.98, 95% CI 0.83 to 1.15) or improved endometrial cancer-related survival (time-to-event data: five trials, 2965 women; HR 0.96, 95% CI 0.72 to 1.28; and dichotomous data: seven trials, 3628 women; RR 1.02, 95% CI 0.81 to 1.29) or improved distant recurrence rates (dichotomous data: seven trials, 3628 women; RR 1.04, 95% CI 0.80 to 1.35).EBRT did not improve survival outcomes in either the intermediate-risk or high-risk subgroups, although high-risk data were limited, and a benefit of EBRT for high-risk women could not be excluded. One trial (PORTEC-2) compared EBRT with VBT in the high-intermediate risk group and reported that VBT was effective in ensuring vaginal control with a non-significant difference in loco-regional relapse rate compared to EBRT (5.1% versus 2.1%; HR 2.08, 95% CI 0.71 to 6.09; P = 0.17). In the subgroup of low-risk patients (IA/B and grade 1/2), EBRT increased the risk of endometrial carcinoma-related deaths (including treatment-related deaths) (two trials, 517 women; RR 2.64, 95% CI 1.05 to 6.66) but there was a lack of data on overall survival. We considered the evidence for the low-risk subgroup to be of a low quality.EBRT was associated with significantly increased severe acute toxicity (two trials, 1328 patients, RR 4.68, 95% CI 1.35 to 16.16), increased severe late toxicity (six trials, 3501 women; RR 2.58, 95% CI 1.61 to 4.11) and significant reductions in quality of life scores and rectal and bladder function more than 10 years after randomisation (one trial, 351 women) compared with no EBRT.One trial of VBT versus no additional treatment in women with low-risk lesions reported a non-significant reduction in locoregional recurrence in the VBT group compared with the no additional treatment group (RR 0.39, (95% CI 0.14 to 1.09). There were no significant differences in survival outcomes in this trial. AUTHORS' CONCLUSIONS EBRT reduces the risk of locoregional recurrence but has no significant impact on cancer-related deaths or overall survival. It is associated with significant morbidity and a reduction in quality of life. There is no demonstrable survival advantage from adjuvant EBRT for high-risk stage I endometrial cancer, however, the meta-analyses of this subgroup were underpowered and also included high-intermediate risk women, therefore we cannot exclude a small benefit in the high-risk subgroup. EBRT may have an adverse effect on endometrial cancer survival when used to treat uncomplicated low-risk (IA/B grade 1/2) endometrial cancer. For the intermediate to high-intermediate risk group, VBT alone appears to be adequate in ensuring vaginal control compared to EBRT. Further research is needed to guide practice for lesions that are truly high risk. In addition, the definitions of risk should be standardised.
Collapse
Affiliation(s)
- Anthony Kong
- Department of Oncology, Oxford University Hospitals NHS Trust and Oxford University, Oxford, UK.
| | | | | | | |
Collapse
|
31
|
Fatima I, Chandra V, Saxena R, Manohar M, Sanghani Y, Hajela K, Negi MPS, Sankhwar PL, Jain SK, Dwivedi A. 2,3-Diaryl-2H-1-benzopyran derivatives interfere with classical and non-classical estrogen receptor signaling pathways, inhibit Akt activation and induce apoptosis in human endometrial cancer cells. Mol Cell Endocrinol 2012; 348:198-210. [PMID: 21878365 DOI: 10.1016/j.mce.2011.08.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 08/11/2011] [Accepted: 08/12/2011] [Indexed: 02/03/2023]
Abstract
OBJECTIVES The present study was undertaken to explore the mechanism of anti-proliferative action of benzopyran compound D1 (2-[piperidinoethoxyphenyl]-3-phenyl-2H-benzopyran) and its hydroxy-(D2) and methoxy-(D3) derivatives in Ishikawa and human primary endometrial adenocarcinoma cells. METHODS Transcriptional activation assays were performed using luciferase reporter system and cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). The stage of cell cycle was determined by flow-cytometry and real time analysis of cyclinE1 and cdc2 genes. The apoptotic effects were measured by AnnexinV/PI staining and TUNEL. The expression of PCNA, cyclinD1, pAkt, XIAP, cleaved caspase-9, -3, PARP, Bax and Bcl2 were determined by immunoblotting. The caspase-3 activity and mitochondrial membrane potential were measured by colorimetric assay. RESULTS All three compounds inhibited E(2)-induced ERE- and AP-1-mediated transactivation and proliferation in endometrial adenocarcinoma cells dose-dependently. Compound D1 caused the arrest of cells in the G(2) phase while D2 and D3 caused arrest in G(1) phase of the cell cycle. All compounds interfered with Akt activation, decreased XIAP expression leading to an increased cleavage of caspase-9, -3, PARP, increased Bax/Bcl2 ratio and caspase-3 activity. CONCLUSION Findings suggest that benzopyran derivatives inhibit cellular proliferation via modulating ER-dependent classical and non-classical signaling mechanisms, interfere with Akt activation and induce apoptosis via intrinsic pathway in endometrial adenocarcinoma cells.
Collapse
Affiliation(s)
- I Fatima
- Division of Endocrinology, Central Drug Research Institute, CSIR, Lucknow 226001, UP, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
The role of the phosphatidylinositol 3-kinase (PI3K) pathway in the development and treatment of uterine cancer. Gynecol Oncol 2011; 123:411-20. [DOI: 10.1016/j.ygyno.2011.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 08/03/2011] [Accepted: 08/03/2011] [Indexed: 11/19/2022]
|
33
|
Marchetti C, Pisano C, Mangili G, Lorusso D, Panici PB, Silvestro G, Candiani M, Greggi S, Perniola G, Di Maio M, Pignata S. Use of adjuvant therapy in patients with FIGO stage III endometrial carcinoma: a multicenter retrospective study. Oncology 2011; 81:104-12. [PMID: 21986449 DOI: 10.1159/000331677] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 08/05/2011] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Adjuvant treatment for stage III endometrial cancer is not yet defined. Previous experiences support the usefulness of combined chemotherapy and radiotherapy. The aim of this retrospective study was to describe the outcome in a cohort of patients with stage III endometrial cancer treated with chemotherapy and/or radiotherapy. METHODS A multicenter retrospective analysis of patients with stage III endometrial cancer from 1998 to 2009 was conducted. The impact on relapse-free survival of clinical and pathological variables and adjuvant treatment received was analyzed by univariate and multivariate analysis. RESULTS Eighty-two patients were considered. Median age was 62 years (range 38-82). Seventy-eight (95%) patients received an adjuvant treatment: chemotherapy (41; 50%), radiotherapy (18; 22%), or combined chemo-radiotherapy (19; 23%). Four patients were excluded from analysis because they were not treated with any adjuvant therapy. At univariate analysis, tumor grade (G3 vs. G1-G2; p = 0.003) was associated with risk of recurrence; similarly, patients treated with radiotherapy alone (p = 0.031, hazard ratio 0.19, 95% CI 0.04-0.86) or chemotherapy alone (p = 0.053, hazard ratio 0.54, 95% CI 0.29-1.01) had a significantly higher risk for relapse, compared to those treated with the multimodality approach. Relapse-free survival at 3 years was 86.5, 65.8 and 44.1%, with the multimodality approach, chemotherapy and radiotherapy, respectively. At multivariable analysis, age and grading were independently associated with recurrence-free survival. Hazard ratio for relapse-free survival was 0.14 (95% CI 0.02-1.04) and 0.20 (95% CI 0.04-1.11) for multimodality treatment compared to chemotherapy alone and radiotherapy alone, respectively. CONCLUSIONS Age and grading are independent prognostic factors. A combined approach with radiotherapy and chemotherapy may induce an advantage in relapse-free survival compared to radiotherapy or chemotherapy alone. Prospective clinical trials are needed to verify this clinical hypothesis.
Collapse
Affiliation(s)
- Claudia Marchetti
- Dipartimento di Scienze Ginecologico-Ostetriche e Scienze Urologiche, Università di Roma Sapienza, Roma, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wincewicz A, Baltaziak M, Kanczuga-Koda L, Koda M, Sulkowska U, Famulski W, Sulkowski S. STAT3 and apoptosis regulators: Bak and Bcl-xL in endometrioid adenocarcinomas of different estrogen receptor-α immunoprofile. Gynecol Endocrinol 2011; 27:536-40. [PMID: 21726118 DOI: 10.3109/09513590.2010.507286] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Estrogen receptor (ER) is a major feature of endometrioid adenocarcinoma. It has a significant impact on constitution of estrogen-responsiveness of this endometrial malignancy, in which STAT3 (signal transducer and activator of transcription) becomes hyperactivated. The aim of our study was to detect immunohistochemically and compare expressions of STAT3 with apoptosis regulators (Bak and Bcl-xL) in regard to different pathological features and variably pronounced ER-α immunoprofile in 78 endometrioid adenocarcinomas. STAT3 was abundantly detected in nuclei of cancer cells in 54 cases, thus pointing at its activation as an universal nuclear transcriptional factor. Bcl-xL and Bak were expressed in cytoplasm of malignant cells in 62 and 20 cancers, respectively. STAT3 correlated both with Bcl-xL (p = 0.001, r = 0.365) and Bak (p < 0.001, r = 0.436) in all of endometrioid adenocarcinomas and variably in different subgroups of these tumours segregated in regard to grading, staging and patients' age. Remarkably, only ER-α positive cancers retained these correlations in opposition to ER-α negative tumours with negativity defined as an immunoreactivity below 10%. ER-α receptor probably enhances interactions between STAT3 and Bcl-xL to be present in statistically significant manner. Presence of ER-α receptor seems to be crucial for relationships among Bcl-xL and STAT3 to occur in endometrioid adenocarcinomas.
Collapse
Affiliation(s)
- Andrzej Wincewicz
- Department of Pathology, Maria Sklodowska-Curie Memorial Bialystok Oncology Center and Medical University of Bialystok, Ogrodowa 12, 15-027 Bialystok, Poland.
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Cytotoxic therapy and surgery have improved outcomes for patients with gynecologic malignancies over the last twenty years, but women's cancers still account for over ten percent of cancer related deaths annually. Insights into the pathogenesis of cancer have led to the development of drugs that target molecular pathways essential to tumor survival including angiogenesis, DNA repair, and apoptosis. This review outlines several of the promising new biologically targeted drugs currently being tested to treat gynecologic malignancies.
Collapse
Affiliation(s)
- Amy R. Carroll
- Department of Gynecologic Oncology, M.D. Anderson Cancer Center, Houston, TX 77030
| | - Robert L. Coleman
- Department of Gynecologic Oncology, M.D. Anderson Cancer Center, Houston, TX 77030
- Center for RNAi and Non-Coding RNA, M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Anil K. Sood
- Department of Gynecologic Oncology, M.D. Anderson Cancer Center, Houston, TX 77030
- Department of Cancer Biology, M.D. Anderson Cancer Center, Houston, TX 77030
- Center for RNAi and Non-Coding RNA, M.D. Anderson Cancer Center, Houston, TX, 77030
| |
Collapse
|