Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Ann Surg Oncol ; 30(5): 2964-2973, 2023 May.
Article in English | MEDLINE | ID: mdl-36920588

ABSTRACT

PURPOSE: To investigate the clinical practices of diagnosing multicystic cervical lesions as a means to develop a more appropriate diagnostic algorithm for gastric-type adenocarcinoma (GAS) and its precursors. METHODS: Clinical information for 159 surgically treated patients for multicystic disease of the uterine cervix was collected from 15 hospitals. We performed a central review of the MRI and pathological findings. The MRI findings were categorized into four types including two newly proposed imaging features based on the morphology and distribution of cysts, and the diagnosis accuracy was assessed. Among the four MRI types, types 1 and 2 were categorized as benign lesions that included LEGH; type 3 were precancerous lesions (with an assumption of atypical LEGH); and type 4 were malignant lesions. RESULTS: The central pathological review identified 56 cases of LEGH, seven with GAS, four with another form of carcinoma, and 92 with benign disease. In clinical practice, over-diagnosis of malignancy (suspicion of MDA) occurred for 12/19 cases (63.2%) and under-diagnosis of malignancy occurred for 4/11 (36%). Among the 118 patients who had a preoperative MRI and underwent a hysterectomy, type 3 or 4 MRI findings in conjunction with abnormal cytology were positively indicative of premalignancy or malignancy, with a sensitivity and specificity of 61.1% and 96.7%, respectively. CONCLUSIONS: Although the correct preoperative diagnosis of cervical cancer with a multicystic lesion is challenging, the combination of cytology and MRI findings creates a more appropriate diagnostic algorithm that significantly improves the diagnostic accuracy for differentiating benign disease from premalignancy and malignancy.


Subject(s)
Adenocarcinoma , Precancerous Conditions , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/diagnostic imaging , Uterine Cervical Neoplasms/surgery , Cervix Uteri/surgery , Cervix Uteri/pathology , Adenocarcinoma/surgery , Adenocarcinoma/pathology , Precancerous Conditions/diagnosis , Precancerous Conditions/surgery , Precancerous Conditions/pathology , Magnetic Resonance Imaging
3.
Am J Reprod Immunol ; 87(6): e13533, 2022 06.
Article in English | MEDLINE | ID: mdl-35366371

ABSTRACT

PROBLEM: The pathogenesis of endometriosis remains unclear. Endometrial cells in retrograde menstruation are considered the source of endometriosis; therefore, we hypothesized that the eutopic endometrium may provide clues regarding the pathogenesis. We aimed to clarify the role of eutopic endometrial cells in endometriosis development. METHOD OF STUDY: Eutopic endometrial tissues were obtained from patients with or without endometriosis, and expression of cell surface molecules in eutopic endometrial stromal cells (ESCs) was evaluated via iTRAQ-based proteomic analysis. Based on the results, we focused on galectin-3. Galectin-3 expression in clinical samples was confirmed by immunohistochemistry and Western blot analysis. The concentration of secreted galectin-3 was measured using enzyme-linked immunosorbent assays. Adhesion and migration of ESCs were evaluated by in vitro adhesion and wound healing assays. The cytotoxicity of natural killer cells was measured via calcein release assays. Cell proliferation was measured using the CyQUANT Cell Proliferation Assay Kit. RESULTS: iTRAQ analysis revealed that galectin-3 expression was specifically elevated in the ESCs from endometriosis patients. Immunohistochemistry confirmed galectin-3 overexpression in the eutopic endometrium of endometriosis, irrespective of the menstrual phase. Galectin-3 was overexpressed and secreted by the eutopic ESCs from patients with endometriosis compared to that from patients without endometriosis. Galectin-3 expression in ESCs increased adhesion and migration, whereas galectin-3 inhibitors impaired these processes. Galectin-3 reduced the cytotoxicity of natural killer cells toward ESCs, while not affecting cell proliferation. CONCLUSION: Galectin-3 promotes peritoneal engraftment of ESCs due to impaired immune surveillance in the peritoneal cavity and increases ESCs adhesion and migration to the peritoneum.


Subject(s)
Endometriosis , Antigens, Neoplasm , Biomarkers, Tumor , Cell Survival , Endometrium/pathology , Female , Galectin 3/genetics , Galectin 3/metabolism , Humans , Peritoneal Cavity/pathology , Proteomics , Stromal Cells/metabolism
4.
Int J Surg Case Rep ; 69: 1-4, 2020.
Article in English | MEDLINE | ID: mdl-32229423

ABSTRACT

INTRODUCTION: Ovarian malignant lymphoma is a rare gynecologic disease and some patients show marked ascites, similar to that observed in advanced ovarian cancer. Although radical surgery improves prognosis of ovarian cancer, treatment of lymphoma is based on chemotherapy, therefore, differential diagnosis is crucial. PRESENTATION OF CASE: A 65-year-old woman presented with a 1-month history of abdominal distention. Pelvic ultrasonography showed an 11-cm solid mass in the pelvis. Computed tomography and magnetic resonance imaging revealed bilateral (mainly left) ovarian masses in the pelvis and multiple metastases. Laboratory examination revealed that serum CA125 levels were elevated, suggesting the existence of advanced ovarian cancer. To confirm the diagnosis, the ascites was removed via abdominocentesis. Although no malignant epithelial cells were observed, atypical lymphoid cells dispersed in the ascites were detected in the cytological analyses. Thus, for accurate diagnosis, we performed re-abdominocentesis and immunohistochemical (IHC) analysis using cell block technique. Cell block analysis showed negative staining for CD3 and positive staining for CD20 in large atypical lymphoid cells, suggesting the existence of large B-cell lymphoma. Repeat blood examination showed that the serum sIL-2R level was elevated. We decided to perform biopsy to make the final treatment decision. Histologically, the tumor demonstrated diffuse proliferation of large atypical lymphoid cells. IHC analysis showed CD3(-), CD5(+), and CD20(+). In addition, IHC analysis also showed CD79a(+), CD10(-), bcl-2(+), and cyclin D1(-). The final diagnosis was diffuse large B-cell lymphoma. DISCUSSION AND CONCLUSION: Here, we present the case of a patient with ovarian malignant lymphoma that was diagnosed using cell block analysis.

5.
Int J Gynecol Cancer ; 26(4): 610-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26905334

ABSTRACT

OBJECTIVE: Aberrant activation of nuclear factor-kappa ß (NF-κB) signaling has been correlated with poor outcome among patients with ovarian cancer. Although the therapeutic potential of NF-κB pathway disruption in cancers has been extensively studied, most classical NF-κB inhibitors are poorly selective, exhibit off-target effects, and have failed to be applied in clinical use. IMD-0560, N-[2,5-bis (trifluoromethyl) phenyl]-5-bromo-2-hydroxybenzamide, is a novel low-molecular-weight compound that selectively inhibits the IκB kinase complex and works as an inhibitor of NF-κB signaling. The aim of this study was to assess the therapeutic potential of IMD-0560 against ovarian cancer in vitro and in vivo. METHODS: NF-κB activity (phosphorylation) was determined in 9 ovarian cancer cell lines and the inhibitory effect of IMD-0560 on NF-κB activation was analyzed by Western blotting. Cell viability, cell cycle, vascular endothelial growth factor (VEGF) expression, and angiogenesis were assessed in vitro to evaluate the effect of IMD-0560 on ovarian cancer cells. In vivo efficacy of IMD-0560 was also investigated using an ovarian cancer xenograft mouse model. RESULTS: The NF-κB signaling pathway was constitutively activated in 8 of 9 ovarian cancer cell lines. IMD-0560 inhibited NF-κB activation and suppressed ovarian cancer cell proliferation by inducing G1 phase arrest. IMD-0560 decreased VEGF secretion from cancer cells and inhibited the tube formation of human umbilical vein endothelial cells. IMD-0560 significantly inhibited peritoneal metastasis and prolonged the survival in an ovarian cancer xenograft mice model. Immunohistochemical staining of excised tumors revealed that IMD-0560 suppressed VEGF expression, tumor angiogenesis, and cancer cell proliferation. CONCLUSIONS: IMD-0560 showed promising therapeutic efficacy against ovarian cancer xenograft mice by inducing cell cycle arrest and suppressing VEGF production from cancer cells. IMD-0560 may be a potential future option in regimens for the treatment of ovarian cancer.


Subject(s)
Apoptosis/drug effects , Benzamides/pharmacology , Cell Proliferation/drug effects , I-kappa B Kinase/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Animals , Blotting, Western , Cell Cycle/drug effects , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Am J Pathol ; 186(3): 616-29, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26796146

ABSTRACT

Preterm delivery (PTD) remains a serious challenge in perinatology. Intrauterine infection and/or inflammation, followed by increased inflammatory cytokines, represented by IL-6, are involved in this pathology. Our aim was to identify IL-6-producing cells in the placenta and to analyze the potential of targeting IκB kinase ß (IKKß) signaling to suppress IL-6 production for the treatment of PTD. Immunohistochemical analyses using placentas complicated with severe chorioamnionitis revealed that IL-6 is mainly expressed in human amniotic mesenchymal stromal cells (hAMSCs). Primary hAMSCs were collected, and strong IL-6 expression was confirmed. In hAMSCs, the treatment of tumor necrosis factor-α or IL-1ß drastically induced IL-6 production, followed by the phosphorylation of IKKs. A novel IKKß inhibitor, IMD-0560, almost completely inhibited IL-6 production from hAMSCs. Using an experimental lipopolysaccharide-induced PTD mouse model, the therapeutic potential of IMD-0560 was examined. IMD-0560 was delivered vaginally 4 hours before lipopolysaccharide administration. Mice in the IMD-0560 (30 mg/kg, twice a day) group had a significantly lower rate of PTD [10 of 22 (45%)] without any apparent adverse events on the mice and their pups. In uteri collected from mice, IMD-0560 inhibited not only IL-6 production but also production of related cytokines, such as keratinocyte-derived protein chemokine/CXCL1, macrophage inflammatory protein-2/CXCL2, and monocyte chemoattractant protein-1/chemokine ligand 2. Targeting IKKß signaling shows promising effects through the suppression of these cytokines and can be explored as a future option for the prevention of PTD.


Subject(s)
Benzamides/administration & dosage , I-kappa B Kinase/antagonists & inhibitors , Inflammation/complications , Interleukin-6/metabolism , Premature Birth/prevention & control , Amnion/cytology , Amnion/metabolism , Animals , Chorioamnionitis/metabolism , Disease Models, Animal , Female , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Interleukin-6/blood , Interleukin-6/genetics , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred C3H , Placenta/metabolism , Pregnancy , Premature Birth/chemically induced , Premature Birth/etiology
7.
Oncotarget ; 6(13): 11342-56, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25839163

ABSTRACT

During the dissemination of ovarian cancer cells, the cells float in the peritoneal cavity without access to a vascular supply and so are exposed to hypoxic conditions, which may cause the ovarian cancer cells to acquire a more aggressive and malignant phenotype. In this study, we screened microRNAs (miRNAs) to identify those that displayed altered expression patterns under hypoxic conditions and then analyzed their functional roles in ovarian cancer progression. miRNA PCR arrays performed on cells from 2 ovarian cancer cell lines (CaOV3 and RMUG-S) revealed miR-199a-3p as one of the miRNAs that are downregulated under hypoxia. In silico analyses indicated that MET is one of the target genes for miR-199a-3p; subsequently, miR-199a-3p expression was found to be inversely correlated with c-Met expression in ovarian cancer. Transfection of precursor miR-199a-3p into ovarian cancer cells reduced c-Met expression and inhibited the phosphorylation of c-Met, extracellular signal-regulated kinase, and AKT; in addition, proliferation, adhesion, and invasiveness were inhibited. Moreover, overexpression of miR-199a-3p in cancer cells significantly suppressed peritoneal dissemination in a xenograft model. In summary, the hypoxia-related microRNA miR-199a-3p drastically inhibits ovarian cancer progression through the downregulation of c-Met expression. Therefore, miR-199a-3p is a potential target for treating ovarian cancer dissemination.


Subject(s)
Carcinoma/metabolism , MicroRNAs/metabolism , Ovarian Neoplasms/metabolism , Animals , Carcinoma/genetics , Carcinoma/pathology , Cell Adhesion , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Disease Progression , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNAi Therapeutics , Signal Transduction , Time Factors , Transfection , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 14(4): 909-19, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637316

ABSTRACT

The prolongation of progression-free survival (PFS) in patients with advanced ovarian cancer by antiangiogenic therapy has been shown in several clinical trials. However, although an anti-VEGF antibody (bevacizumab) is the only option currently available, its efficacy is limited and it is not cost effective for use in all patients. Therefore, the development of a novel antiangiogenic drug, especially composed of small-molecule compounds, could be a powerful armament for ovarian cancer treatment. As NF-κB signaling has the potential to regulate VEGF expression, we determined to identify whether VEGF expression is associated with NF-κB activation and to investigate the possibility of a novel IKKß inhibitor, IMD-0354 (IMMD Inc.), as an antiangiogenic drug. Tissue microarrays from 94 ovarian cancer tissues were constructed and immunohistochemical analyses performed. We revealed that IKK phosphorylation is an independent prognostic factor (PFS: 26.1 vs. 49.8 months, P = 0.011), and is positively correlated with high VEGF expression. In in vitro analyses, IMD-0354 robustly inhibited adhesive and invasive activities of ovarian cancer cells without impairing cell viabilities. IMD-0354 significantly suppressed VEGF production from cancer cells, which led to the inhibition of angiogenesis. In a xenograft model, the treatment of IMD-0354 significantly inhibited peritoneal dissemination with a marked reduction of intratumoral blood vessel formation followed by the inhibition of VEGF expression from cancer cells. IMD-0354 is a stable small-molecule drug and has already been administered safely to humans in other trials. Antiangiogenic therapy targeting IKKß is a potential future option to treat ovarian cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , I-kappa B Kinase/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Vascular Endothelial Growth Factors/genetics , Animals , Benzamides/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Enzyme Activation , Female , Humans , Immunohistochemistry , Mice , Molecular Targeted Therapy , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Phosphorylation , Prognosis , Vascular Endothelial Growth Factors/antagonists & inhibitors , Vascular Endothelial Growth Factors/metabolism , Xenograft Model Antitumor Assays
9.
PLoS One ; 10(2): e0118080, 2015.
Article in English | MEDLINE | ID: mdl-25658637

ABSTRACT

Ovarian cancer remains the most lethal gynecologic cancer and new targeted molecular therapies against this miserable disease continue to be challenging. In this study, we analyzed the expressional patterns of Interleukin-6 (IL-6) and its receptor (IL-6R) expression in ovarian cancer tissues, evaluated the impact of these expressions on clinical outcomes of patients, and found that a high-level of IL-6R expression but not IL-6 expression in cancer cells is an independent prognostic factor. In in vitro analyses using ovarian cell lines, while six (RMUG-S, RMG-1, OVISE, A2780, SKOV3ip1 and OVCAR-3) of seven overexpressed IL-6R compared with a primary normal ovarian surface epithelium, only two (RMG-1, OVISE) of seven cell lines overexpressed IL-6, suggesting that IL-6/IL-6R signaling exerts in a paracrine manner in certain types of ovarian cancer cells. Ovarian cancer ascites were collected from patients, and we found that primary CD11b+CD14+ cells, which were predominantly M2-polarized macrophages, are the major source of IL-6 production in an ovarian cancer microenvironment. When CD11b+CD14+ cells were co-cultured with cancer cells, both the invasion and the proliferation of cancer cells were robustly promoted and these promotions were almost completely inhibited by pretreatment with anti-IL-6R antibody (tocilizumab). The data presented herein suggest a rationale for anti-IL-6/IL-6R therapy to suppress the peritoneal spread of ovarian cancer, and represent evidence of the therapeutic potential of anti-IL-6R therapy for ovarian cancer treatment.


Subject(s)
Ovarian Neoplasms/metabolism , Receptors, Interleukin-6/metabolism , Adult , Aged , Aged, 80 and over , CD11b Antigen/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression , Humans , Immunophenotyping , Interleukin-6/metabolism , Interleukin-6/pharmacology , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Macrophages/pathology , Middle Aged , Models, Biological , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/drug therapy , Paracrine Communication , Phenotype , Prognosis , Receptors, Interleukin-6/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...