Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Diagn Pathol ; 16(1): 98, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34706741

ABSTRACT

BACKGROUND: Aurora-A kinase is important for cellular proliferation and is implicated in the tumorigenesis of several malignancies, including of the ovary. Information regarding the expression patterns of Aurora-A in normal Müllerian epithelium as well as benign, borderline and malignant epithelial ovarian neoplasms is limited. METHODS: We investigated Aurora-A expression by immunohistochemistry in 15 benign, 19 borderline and 17 malignant ovarian serous tumors, and 16 benign, 8 borderline, and 2 malignant ovarian mucinous tumors. Twelve fimbriae from seven patients served as normal Müllerian epithelium controls. We also examined Aurora-A protein expression by western blot in normal fimbriae and tumor specimens. RESULTS: All normal fimbriae (n = 12) showed nuclear but not cytoplasmic Aurora-A immunoreactivity by immunohistochemistry. Benign ovarian tumors also showed strong nuclear Aurora-A immunoreactivity. Forty-eight percent (13/27) of borderline tumors demonstrated nuclear Aurora-A immunoreactivity, while the remainder (52%, 14/27) lacked Aurora-A staining. Nuclear Aurora-A immunoreactivity was absent in all malignant serous tumors, however, 47% (8/17) demonstrated perinuclear cytoplasmic staining. These results were statistically significant when tumor class (benign/borderline/malignant) was compared to immunoreactivity localization or intensity (Fisher Exact Test, p < 0.01). Western blot analysis confirmed the greater nuclear Aurora-A expression in control Müllerian epithelium compared to borderline and malignant tumors. CONCLUSION: Aurora-A kinase is differentially expressed across normal Müllerian epithelium, benign and borderline serous and mucinous ovarian epithelial neoplasms and malignant serous ovarian tumors., with nuclear expression of unphosphorylated Aurora-A being present in normal and benign neoplastic epithelium, and lost in malignant serous neoplasms. Further studies of the possible biological and clinical implications of the loss of nuclear Aurora-A expression in ovarian tumors, and its role in ovarian carcinogenesis are warranted.


Subject(s)
Aurora Kinase A/biosynthesis , Carcinoma, Ovarian Epithelial/enzymology , Cystadenocarcinoma, Mucinous/enzymology , Cystadenocarcinoma, Serous/enzymology , Ovary/enzymology , Carcinoma, Ovarian Epithelial/pathology , Cell Nucleus/enzymology , Cystadenocarcinoma, Mucinous/pathology , Cystadenocarcinoma, Serous/pathology , Cytoplasm/enzymology , Epithelium/enzymology , Female , Humans
2.
Oncogene ; 40(44): 6248-6257, 2021 11.
Article in English | MEDLINE | ID: mdl-34556811

ABSTRACT

The enzyme iodothyronine deiodinase type 3 (DIO3) contributes to cancer proliferation by inactivating the tumor-suppressive actions of thyroid hormone (T3). We recently established DIO3 involvement in the progression of high-grade serous ovarian cancer (HGSOC). Here we provide a link between high DIO3 expression and lower survival in patients, similar to common disease markers such as Ki67, PAX8, CA-125, and CCNE1. These observations suggest that DIO3 is a logical target for inhibition. Using a DIO3 mimic, we developed original DIO3 inhibitors that contain a core of dibromomaleic anhydride (DBRMD) as scaffold. Two compounds, PBENZ-DBRMD and ITYR-DBRMD, demonstrated attenuated cell counts, induction in apoptosis, and a reduction in cell proliferation in DIO3-positive HGSOC cells (OVCAR3 and KURAMOCHI), but not in DIO3-negative normal ovary cells (CHOK1) and OVCAR3 depleted for DIO3 or its substrate, T3. Potent tumor inhibition with a high safety profile was further established in HGSOC xenograft model, with no effect in DIO3-depleted tumors. The antitumor effects are mediated by downregulation in an array of pro-cancerous proteins, the majority of which known to be repressed by T3. To conclude, using small molecules that specifically target the DIO3 enzyme we present a new treatment paradigm for ovarian cancer and potentially other DIO3-dependent malignancies.


Subject(s)
Carcinoma, Ovarian Epithelial/drug therapy , Cystadenocarcinoma, Serous/drug therapy , Enzyme Inhibitors/administration & dosage , Iodide Peroxidase/metabolism , Small Molecule Libraries/administration & dosage , Animals , Carcinoma, Ovarian Epithelial/enzymology , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Down-Regulation , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Iodide Peroxidase/antagonists & inhibitors , Iodide Peroxidase/genetics , Mice , Molecular Mimicry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays
3.
Am J Pathol ; 191(10): 1837-1850, 2021 10.
Article in English | MEDLINE | ID: mdl-34214505

ABSTRACT

Deregulated full-length anaplastic lymphoma kinase (ALK) overexpression has been found in some primary solid tumors, but little is known about its role in ovarian high-grade serous carcinoma (HGSC). The current study focused on the functional roles of ALK in HGSC. Cytoplasmic ALK immunoreactivity without chromosomal rearrangement and gene mutations was significantly higher in HGSC compared with non-HGSC-type ovarian carcinomas, and was significantly associated with several unfavorable clinicopathologic factors and poor prognosis. HGSC cell lines stably overexpressing ALK exhibited increased cell proliferation, enhanced cancer stem cell features, and accelerated cell mobility, whereas these phenotypes were abrogated in ALK-knockdown cells. Expression of the nervous system-associated gene, ELAVL3, and the corresponding protein (commonly known as HuC) was significantly increased in cells overexpressing ALK. Expression of SRY-box transcription factor (Sox)2 and Sox3 (genes associated with the neural progenitor population) increased in ALK-overexpressing but not ALK-knockdown cells. Furthermore, overexpression of Sox2 or Sox3 enhanced both ALK and ELAVL3 promoter activities, suggesting the existence of ALK/Sox/HuC signaling loops. Finally, ALK overexpression was attributed to increased expression of neuroendocrine markers, including synaptophysin, CD56, and B-cell lymphoma 2, in HGSC tissues. These findings suggest that overexpression of full-length ALK may influence the biological behavior of HGSC through cooperation with ELAVL3 and Sox factors, leading to the establishment and maintenance of the aggressive phenotypic characteristics of HGSC.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Adult , Aged , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cytoplasm/enzymology , ELAV-Like Protein 3/metabolism , Female , Humans , Middle Aged , Models, Biological , Multivariate Analysis , Neoplasm Grading , Neoplastic Stem Cells/pathology , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Phenotype , Prognosis , Progression-Free Survival , SOX Transcription Factors/metabolism
4.
J Clin Oncol ; 38(32): 3753-3762, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32822286

ABSTRACT

PURPOSE: Low-grade serous ovarian carcinomas (LGSOCs) have historically low chemotherapy responses. Alterations affecting the MAPK pathway, most commonly KRAS/BRAF, are present in 30%-60% of LGSOCs. The purpose of this study was to evaluate binimetinib, a potent MEK1/2 inhibitor with demonstrated activity across multiple cancers, in LGSOC. METHODS: This was a 2:1 randomized study of binimetinib (45 mg twice daily) versus physician's choice chemotherapy (PCC). Eligible patients had recurrent measurable LGSOC after ≥ 1 prior platinum-based chemotherapy but ≤ 3 prior chemotherapy lines. The primary end point was progression-free survival (PFS) by blinded independent central review (BICR); additional assessments included overall survival (OS), overall response rate (ORR), duration of response (DOR), clinical-benefit rate, biomarkers, and safety. RESULTS: A total of 303 patients were randomly assigned to an arm of the study at the time of interim analysis (January 20, 2016). Median PFS by BICR was 9.1 months (95% CI, 7.3 to 11.3) for binimetinib and 10.6 months (95% CI, 9.2 to 14.5) for PCC (hazard ratio,1.21; 95%CI, 0.79 to 1.86), resulting in early study closure according to a prespecified futility boundary after 341 patients had enrolled. Secondary efficacy end points were similar in the two groups: ORR 16% (complete response [CR]/partial responses[PRs], 32) versus 13% (CR/PRs, 13); median DOR, 8.1 months (range, 0.03 to ≥ 12.0 months) versus 6.7 months (0.03 to ≥ 9.7 months); and median OS, 25.3 versus 20.8 months for binimetinib and PCC, respectively. Safety results were consistent with the known safety profile of binimetinib; the most common grade ≥ 3 event was increased blood creatine kinase level (26%). Post hoc analysis suggests a possible association between KRAS mutation and response to binimetinib. Results from an updated analysis (n = 341; January 2019) were consistent. CONCLUSION: Although the MEK Inhibitor in Low-Grade Serous Ovarian Cancer Study did not meet its primary end point, binimetinib showed activity in LGSOC across the efficacy end points evaluated. A higher response to chemotherapy than expected was observed and KRAS mutation might predict response to binimetinib.


Subject(s)
Benzimidazoles/therapeutic use , Cystadenocarcinoma, Serous/drug therapy , Fallopian Tube Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Benzimidazoles/adverse effects , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Fallopian Tube Neoplasms/enzymology , Fallopian Tube Neoplasms/pathology , Female , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Paclitaxel/therapeutic use , Peritoneal Neoplasms/enzymology , Peritoneal Neoplasms/pathology , Polyethylene Glycols/therapeutic use , Progression-Free Survival , Protein Kinase Inhibitors/adverse effects , Topotecan/therapeutic use , Young Adult
5.
Oncogene ; 39(33): 5520-5535, 2020 08.
Article in English | MEDLINE | ID: mdl-32647134

ABSTRACT

High grade serous ovarian cancer (HGSOC) is a fatal gynecologic malignancy in the U.S. with limited treatment options. New therapeutic strategies include targeting of the cell cycle checkpoints, e.g., ATR and CHK1. We recently reported a promising clinical activity of the CHK1 inhibitor (CHK1i) prexasertib monotherapy in BRCA wild-type (BRCAwt) HGSOC patients. In this study, biopsies of treated patients and cell line models were used to investigate possible mechanisms of resistance to CHK1i. We report that BRCAwt HGSOC develops resistance to prexasertib monotherapy via a prolonged G2 delay induced by lower CDK1/CyclinB1 activity, thus preventing cells from mitotic catastrophe and cell death. On the other hand, we noted CHK1's regulation on RAD51-mediated homologous recombination (HR) repair was not altered in CHK1i-resistant cells. Therefore, CHK1i sensitizes CHK1i-resistant cells to DNA damaging agents such as gemcitabine or hydroxyurea by inhibition of HR. In summary, our results demonstrate new mechanistic insights of functionally distinct CHK1 activities and highlight a potential combination treatment approach to overcome CHK1i resistance in BRCAwt HGSOC.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Pyrazines/pharmacology , Pyrazoles/pharmacology , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/genetics , Drug Resistance, Neoplasm , Female , Humans , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics
6.
Cells ; 9(2)2020 02 14.
Article in English | MEDLINE | ID: mdl-32075097

ABSTRACT

Identifying cancer drivers and actionable mutations is critical for precision oncology. In epithelial ovarian cancer (EOC) the majority of mutations lack biological or clinical validation. We fully characterized 43 lines of Patient-Derived Xenografts (PDXs) and performed copy number analysis and whole exome sequencing of 12 lines derived from naïve, high grade EOCs. Pyrosequencing allowed quantifying mutations in the source tumours. Drug response was assayed on PDX Derived Tumour Cells (PDTCs) and in vivo on PDXs. We identified a PIK3R1W624R variant in PDXs from a high grade serous EOC. Allele frequencies of PIK3R1W624R in all the passaged PDXs and in samples of the source tumour suggested that it was truncal and thus possibly a driver mutation. After inconclusive results in silico analyses, PDTCs and PDXs allowed the showing actionability of PIK3R1W624R and addiction of PIK3R1W624R carrying cells to inhibitors of the PI3K/AKT/mTOR pathway. It is noteworthy that PIK3R1 encodes the p85α regulatory subunit of PI3K, that is very rarely mutated in EOC. The PIK3R1W624R mutation is located in the cSH2 domain of the p85α that has never been involved in oncogenesis. These data show that patient-derived models are irreplaceable in their role of unveiling unpredicted driver and actionable variants in advanced ovarian cancer.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Cystadenocarcinoma, Serous/genetics , Ovarian Neoplasms/genetics , Animals , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/enzymology , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/enzymology , Female , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Mutation , Neoplasm Grading , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Random Allocation
8.
Sci Rep ; 9(1): 11677, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406154

ABSTRACT

Ovarian cancer has a high death rate and is often not detected until late stages. While some studies found high expressions of MMPs correlated with cancer invasion, metastasis, and poor prognosis, however, several other studies indicated MMPs might inhibit cancer rather than promote cancer in certain situations. Thus, the role of different MMPs in different cancer types needs a systematic re-evaluation. In this study, we used RNA-Seq and corresponding clinical data downloaded from TCGA and analyzed the correlations between MMP expressions and the clinicopathologic characteristics and outcome in ovarian serous cystadenocarcinoma (OSC) patients. Among the MMPs investigated, MMP-3 was significantly increased in high-grade and high-stage tumors compared with low-grade and low-stage ones. Using univariate analysis and multivariate Cox model, high expressions of MMP-19 and -20 were found to associate with poor overall survival independent of clinicopathologic characteristics. Moreover, using in vitro studies, we found ovarian cancer cell lines with higher MMP-19 and -20 protein expressing levels were associated with anti-cancer drugs resistance, while knockdown of MMP-19 or -20 increased ovarian cancer cell sensitivities to several clinical using chemotherapy agents. Finally, knockdown of MMP-19 or -20 also decreased the invasion abilities of several ovarian cancer cell lines. These in vitro studies provided potential mechanisms of high MMP-19 and -20 expressions in the poor prognosis of ovarian cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Ovarian Epithelial/genetics , Cystadenocarcinoma, Serous/genetics , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinases/genetics , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Asian People , Black People , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/enzymology , Carcinoma, Ovarian Epithelial/mortality , Cell Line, Tumor , Cohort Studies , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/mortality , Female , Humans , Matrix Metalloproteinases/metabolism , Middle Aged , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/mortality , Prognosis , Proportional Hazards Models , White People
9.
Int J Mol Sci ; 20(12)2019 Jun 22.
Article in English | MEDLINE | ID: mdl-31234549

ABSTRACT

High grade serous ovarian cancer (HGSOC) is the most common and aggressive ovarian cancer subtype with the worst clinical outcome due to intrinsic or acquired drug resistance. Standard treatment involves platinum compounds. Cancer development and chemoresistance is often associated with an increase in histone deacetylase (HDAC) activity. The purpose of this study was to examine the potential of HDAC inhibitors (HDACi) to increase platinum potency in HGSOC. Four HGSOC cell lines with different cisplatin sensitivity were treated with combinations of cisplatin and entinostat (class I HDACi), panobinostat (pan-HDACi), or nexturastat A (class IIb HDACi), respectively. Inhibition of class I HDACs by entinostat turned out superior in increasing cisplatin potency than pan-HDAC inhibition in cell viability assays (MTT), apoptosis induction (subG1), and caspase 3/7 activation. Entinostat was synergistic with cisplatin in all cell lines in MTT and caspase activation assays. MTT assays gave combination indices (CI values) < 0.9 indicating synergism. The effect of HDAC inhibitors could be attributed to the upregulation of pro-apoptotic genes (CDNK1A, APAF1, PUMA, BAK1) and downregulation of survivin. In conclusion, the combination of entinostat and cisplatin is synergistic in HGSOC and could be an effective strategy for the treatment of aggressive ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/therapeutic use , Cystadenocarcinoma, Serous/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Ovarian Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Benzamides/pharmacology , Benzamides/therapeutic use , Cell Line, Tumor , Cell Survival , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/physiopathology , Drug Synergism , Female , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/physiopathology , Panobinostat/pharmacology , Panobinostat/therapeutic use , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use
10.
Gynecol Oncol ; 152(2): 408-415, 2019 02.
Article in English | MEDLINE | ID: mdl-30448261

ABSTRACT

OBJECTIVE: The objective of this study was to analyze the expression level and clinical role of soluble AXL (sAXL) in cancers affecting the serosal surfaces, with focus on ovarian carcinoma. METHODS: sAXL protein expression by ELISA was analyzed in 572 effusion supernatants, including 424 peritoneal, 147 pleural and 1 pericardial specimens. RESULTS: sAXL was overexpressed in peritoneal effusions compared to pleural and pericardial specimens (p < 0.001). sAXL levels were additionally significantly higher in effusions from patients with ovarian carcinoma, malignant mesothelioma and breast carcinoma compared to specimens from patients with other cancers (predominantly carcinomas of lung, gastrointestinal or uterine corpus/cervix origin) or benign reactive effusions (p < 0.001). sAXL was further overexpressed in high-grade serous carcinoma (HGSC; n = 373) compared to low-grade serous carcinoma (LGSC; n = 32; p = 0.036). In HGSC, sAXL levels were significantly lower in post-chemotherapy effusions compared to primary diagnosis pre-chemotherapy specimens (p = 0.002). sAXL levels in HGSC were unrelated to chemoresponse at diagnosis, progression-free survival or overall survival. Levels were similarly unrelated to survival in LGSC and breast carcinoma. CONCLUSIONS: sAXL is widely expressed in malignant effusions, particularly in ovarian and breast carcinoma and in malignant mesothelioma. sAXL is overexpressed in HGSC compared to LGSC and its levels are lower following exposure to chemotherapy. However, sAXL levels are not informative of chemoresponse or survival.


Subject(s)
Ascitic Fluid/enzymology , Breast Neoplasms/enzymology , Cystadenocarcinoma, Serous/enzymology , Lung Neoplasms/enzymology , Mesothelioma/enzymology , Ovarian Neoplasms/enzymology , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Adult , Aged , Aged, 80 and over , Ascitic Fluid/drug effects , Ascitic Fluid/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/pathology , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mesothelioma/drug therapy , Mesothelioma/pathology , Mesothelioma, Malignant , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Young Adult , Axl Receptor Tyrosine Kinase
11.
Future Oncol ; 14(25): 2579-2588, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29783891

ABSTRACT

AIM: To explore the expression profile of some DHRS genes in high-grade serous ovarian cancer (SOVC) and to study their prognostic values. PATIENTS & METHODS: A retrospective bioinformatic analysis was performed using data in the Gene Expression Omnibus, the Human Protein Atlas and the Cancer Genome Atlas-Ovarian Cancer. RESULTS: Increased DHRS12 expression was an independent indicator of poor overall survival (hazard ratio [HR]: 1.265, 95% CI: 1.075-1.488; p = 0.005) and recurrence-free survival (RFS; HR: 2.242, 95%CI: 1.464-3.432; p < 0.001) in patients with high-grade SOVC. DNA deletion was associated with decreased DHRS12 expression, as well as the best overall survival and RFS among the three copy number alteration groups. CONCLUSION: DHRS12 might serve as a valuable prognostic biomarker in high-grade SOVC.


Subject(s)
Alcohol Oxidoreductases/analysis , Cystadenocarcinoma, Serous/mortality , Nuclear Proteins/analysis , Ovarian Neoplasms/mortality , Adult , Aged , Biomarkers, Tumor/analysis , Carbonyl Reductase (NADPH) , Computational Biology , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Female , Humans , Middle Aged , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Prognosis , Retrospective Studies , Transcriptome
12.
Gynecol Oncol ; 150(1): 136-142, 2018 07.
Article in English | MEDLINE | ID: mdl-29804637

ABSTRACT

OBJECTIVE: To analyze the expression and clinical role of CHK1 and CHK2 in metastatic high-grade serous carcinoma (HGSC). METHODS: HGSC effusions (n = 335; 280 peritoneal, 55 pleural) were analyzed for protein expression of total CHK1 and its phosphorylated forms p-ser317 and p-ser296, as well as total CHK2 and its phosphorylated form p-thr68 using immunohistochemistry. Expression was analyzed for association with clinicopathologic parameters, including chemotherapy response, and survival. RESULTS: Carcinoma cells stained positive, predominantly at the nuclei, in the majority of cases (range 83-100% for the five antibodies), while expression in reactive mesothelial cells and tumor-associated macrophages was more variable. Total CHK1 (p = 0.037), p-CHK1ser317 (p = 0.001), p-CHK1ser296 (p = 0.002) and p-CHK2thr68 (p < 0.001) expression was significantly higher in post-chemotherapy disease recurrence compared to pre-chemotherapy effusions obtained at diagnosis. CHK1, p-CHK1ser296, p-CHK2thr68 and p-CHK1ser317 nuclear expression was positively related to expression of the checkpoint regulator WEE1, previously studied in this cohort (p = 0.003, p = 0.013, p = 0.001 and p = 0.01, respectively). Higher total CHK1 (p = 0.007), p-CHK1ser317 (p = 0.004), CHK2 (p = 0.01) and p-CHK2thr68 (p = 0.048) expression was significantly related to shorter overall survival in univariate analysis, and CHK1ser317 was an independent prognostic marker in multivariate analysis (p = 0.025). Higher p-CHK1ser317 (p = 0.03) and CHK2 (p = 0.034) expression was additionally associated with poor progression-free survival. CONCLUSIONS: CHK1 and CHK2 and their activated forms are frequently expressed in HGSC effusions, with higher expression following exposure to chemotherapy, and their expression is related to survival.


Subject(s)
Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/biosynthesis , Checkpoint Kinase 2/metabolism , Cystadenocarcinoma, Serous/enzymology , Adult , Aged , Aged, 80 and over , Checkpoint Kinase 1/biosynthesis , Checkpoint Kinase 1/genetics , Checkpoint Kinase 2/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Enzyme Activation , Female , Humans , Middle Aged , Neoplasm Grading , Survival Analysis , Young Adult
13.
J Cancer Res Clin Oncol ; 144(6): 1109-1118, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29546479

ABSTRACT

PURPOSE: Gene expression of a variety of the 15 members of the KLK serine protease family is dysregulated in ovarian cancer. We aimed at determining the clinical relevance of KLK13 and KLK14 mRNA expression in tumor tissues of a homogeneous patient cohort afflicted with advanced high-grade serous ovarian cancer (FIGO stage III/IV). METHODS: mRNA expression levels of KLK13 and KLK14 were assessed by quantitative PCR in tumor tissue of 91 patients and related with clinical factors and patients' outcome. RESULTS: There was no significant association of KLK13 and KLK14 mRNA expression with the clinical factors ascitic fluid volume or residual tumor mass. In univariate Cox regression analysis, elevated KLK13 mRNA levels were significantly linked with shorter progression-free (PFS; hazard ratio [HR] = 1.97, P = 0.020) and overall survival (OS; HR = 1.81, P = 0.041). High KLK14 mRNA levels were significantly associated with prolonged PFS (HR = 0.44, P = 0.017) and showed a trend towards significance for OS (HR = 0.55, P = 0.070). In multivariable analysis, including the factors age, residual tumor mass, ascitic fluid volume, KLK13, and KLK14, both KLKs, apart from residual tumor mass, remained statistically independent predictive markers: patients with high KLK13 mRNA expression levels displayed a more than twofold increase risk for shorter PFS (HR = 2.14, P = 0.020) as well as OS (HR = 2.05, P = 0.028), whereas elevated KLK14 mRNA values were found to be significant for both, prolonged PFS (HR = 0.36, P = 0.007) and OS (HR = 0.46, P = 0.037). CONCLUSION: These results indicate that in advanced high-grade serous ovarian cancer KLK13 may become proficient for tumor-supporting functions, whereas KLK14 may have adopted tumor-suppressing activity.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Kallikreins/genetics , Ovarian Neoplasms/genetics , RNA, Messenger/biosynthesis , Adult , Age Factors , Aged , Aged, 80 and over , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Disease-Free Survival , Female , Humans , Kallikreins/biosynthesis , Middle Aged , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Survival Rate
14.
J Cell Biochem ; 119(7): 6008-6015, 2018 07.
Article in English | MEDLINE | ID: mdl-29575012

ABSTRACT

Glutamate-ammonia ligase (GLUL), which is also called GS (glutamine synthetase), is the enzyme that catalyzes the synthesis of glutamine from glutamate and ammonia in an ATP-dependent reaction. Here, we found higher expression of GLUL in the ovarian cancer patients was associated with worse disease-free survival (DFS) and overall survival (OS). In addition, GLUL was heterogeneously expressed in various ovarian cancer cells. The mRNA and protein expression levels of GLUL in NIH:OVCAR-3 and ES-2 cells were obviously higher than that in the other types of ovarian cancer cells. Knockdown of GLUL in NIH:OVCAR-3 or ES-2 cells could significantly decrease the proliferation ability. Furthermore, GLUL knockdown markedly inhibited the p38 MAPK signaling pathway in NIH:OVCAR-3 or ES-2 cells. Our findings suggest that decreasing expression of GLUL may be a new approach that can be used for ovarian cancer treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Glutamate-Ammonia Ligase/metabolism , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/enzymology , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Mucinous/enzymology , Adenocarcinoma, Mucinous/pathology , Cell Proliferation , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/pathology , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Invasiveness , Ovarian Neoplasms/enzymology , Prognosis , Survival Rate , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/metabolism
15.
J Clin Oncol ; 36(20): 2044-2051, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29584549

ABSTRACT

Purpose Uterine serous carcinoma is a rare, aggressive variant of endometrial cancer. Trastuzumab is a humanized monoclonal antibody that targets human epidermal growth factor receptor 2 (HER2)/neu, a receptor overexpressed in 30% of uterine serous carcinoma. This multicenter, randomized phase II trial compared carboplatin-paclitaxel with and without trastuzumab in patients with advanced or recurrent uterine serous carcinoma who overexpress HER2/neu. Methods Eligible patients had primary stage III or IV or recurrent HER2/neu-positive disease. Participants were randomly assigned to receive carboplatin-paclitaxel (control arm) for six cycles with or without intravenous trastuzumab (experimental arm) until progression or unacceptable toxicity. The primary end point was progression-free survival, which was assessed for differences between treatment arms via one-sided log-rank tests. Results From August 2011 to March 2017, 61 patients were randomly assigned. Forty progression-free survival-related events occurred among 58 evaluable participants. Among all patients, median progression-free survival was 8.0 months (control) versus 12.6 months (experimental; P = .005; hazard ratio [HR], 0.44; 90% CI, 0.26 to 0.76). Similarly, median progression-free survival was 9.3 (control) versus 17.9 (experimental) months among 41 patients with stage III or IV disease undergoing primary treatment ( P = .013; HR, 0.40; 90% CI, 0.20 to 0.80) and 6.0 (control) versus 9.2 months (experimental), respectively, among 17 patients with recurrent disease ( P = .003; HR, 0.14; 90% CI, 0.04 to 0.53). Toxicity was not different between treatment arms, and no unexpected safety signals emerged. Conclusion Addition of trastuzumab to carboplatin-paclitaxel was well tolerated and increased progression-free survival. These encouraging results deserve further investigation to determine their impact on overall survival in patients with advanced or recurrent uterine serous carcinoma who overexpress HER2/neu.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cystadenocarcinoma, Serous/drug therapy , Receptor, ErbB-2/biosynthesis , Uterine Neoplasms/drug therapy , Aged , Aged, 80 and over , Carboplatin/administration & dosage , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Female , Humans , Middle Aged , Neoplasm Staging , Paclitaxel/administration & dosage , Progression-Free Survival , Trastuzumab/administration & dosage , Uterine Neoplasms/enzymology , Uterine Neoplasms/pathology
16.
Gynecol Oncol ; 149(1): 163-172, 2018 04.
Article in English | MEDLINE | ID: mdl-29429592

ABSTRACT

OBJECTIVE: High grade serous ovarian cancer (HGSC) remains one of the most lethal malignancies in females. We previously reported that γ-glutamyl cyclotransferase (GGCT) was significantly upregulated in serous ovarian cancer. The current study was aimed to explore the function and underlying mechanism of GGCT in HGSC. METHODS: GGCT expression was assessed by immunohistochemistry in 128 HGSC patients. Stable cell lines with GGCT gene overexpression or knockdown were established to investigate the function of GGCT in HGSC in vitro and in vivo. RESULTS: GGCT is highly upregulated in HGSC tissues and associated with FIGO stage, lymph node metastasis and ascitic fluid volume. High expression of GGCT is associated with poor survival in HGSC patients. The Harrell's c-indexes of the prognostic models for overall survival and progression-free survival prediction were 0.758 and 0.726, respectively. GGCT knockdown suppresses proliferation, clone formation, migration, and invasion of tumor cells in vitro while forced GGCT overexpression presents opposite results. Furthermore, GGCT silencing inhibits tumor growth and spread in vivo. Epithelial-mesenchymal transition (EMT) and PI3K/AKT/mTOR signaling pathway are suppressed in GGCT silenced cells and enhanced in GGCT overexpressed cells. Inactivation of PI3K/AKT/mTOR signaling pathway in GGCT overexpressed cells induces EMT inhibition. CONCLUSIONS: Our data reveals an important role of GGCT in regulating EMT and progression of HGSC, providing a valuable prognostic marker and potential target for treatment of HGSC patients.


Subject(s)
Cystadenocarcinoma, Serous/enzymology , Ovarian Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , gamma-Glutamylcyclotransferase/biosynthesis , Animals , Cell Proliferation/physiology , Cystadenocarcinoma, Serous/pathology , Disease Progression , Epithelial-Mesenchymal Transition , Female , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Metastasis , Ovarian Neoplasms/pathology , Signal Transduction , Up-Regulation , gamma-Glutamylcyclotransferase/metabolism
17.
Int J Oncol ; 51(6): 1887-1897, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039611

ABSTRACT

Protein glycosylation perturbations are implicated in a variety of diseases, including cancer. Aberrant glycosylation in cancer is frequently attributed to altered expression of polypeptide GalNAc transferases (GalNAc­Ts) - enzymes initiating mucin-type O-glycosylation. A previous study from our group demonstrated that one member of this family (GALNT3) is overexpressed in epithelial ovarian cancer (EOC), and GALNT3 expression correlated with shorter progression-free survival (PFS) in EOC patients with advanced disease. As considerable degree of redundancy between members of the GalNAc­Ts gene family has been frequently observed, we decided to investigate whether other members of this family are essential in EOC progression. In silico analysis based on publically available data was indicative for altered expression of five GalNAc­Ts (GALNT2, T4, T6, T9 and T14) in ovarian high-grade serous carcinoma (HGSC) samples compared to non-tumoral (control) ovarian tissue. We analyzed protein expression of these GalNAc­Ts in EOC cells and tumors by western blotting, followed by immunohistochemical (IHC) evaluation of their expression in EOC tumor and control samples using tissue microarrays (TMAs). Western blot analyses were indicative for low expression of GALNT2 and strong expression of GALNT6, T9 and T14 in both EOC cells and tumors. These observations were confirmed by IHC. GALNT2 displayed significantly lower expression, while GALNT6, GALNT9 and GALNT14 showed significantly higher expression in HGSC tumors compared to control tissue. Importantly, GALNT6 and GALNT14 expression correlated with poor prognosis of serous EOC patients. Moreover, our results suggest for overlapping functions of some GalNAc­Ts, more specifically GALNT3 and GALNT6, in directing EOC progression. Our results are indicative for a possible implication of different members of the GalNAc­T gene family in modulating EOC progression, and the potential use of GALNT6 and GALNT14 as novel prognostic EOC biomarkers. These data warrant future studies on the role of members of the GalNAc­Ts gene family in ovarian tumorigenesis.


Subject(s)
Cystadenocarcinoma, Serous/enzymology , N-Acetylgalactosaminyltransferases/biosynthesis , Neoplasms, Glandular and Epithelial/enzymology , Ovarian Neoplasms/enzymology , Aged , Biomarkers, Tumor/biosynthesis , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cystadenocarcinoma, Serous/pathology , Disease Progression , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Prognosis , Tissue Array Analysis
18.
IUBMB Life ; 69(10): 802-813, 2017 10.
Article in English | MEDLINE | ID: mdl-28884887

ABSTRACT

We aimed to identify the possible role of oxidant-antioxidant status in epithelial ovarian cancer (EOC) by measuring (a) antioxidant enzyme (AOE) activities [total superoxide dismutase (SODtotal ), manganese-SOD (Mn-SOD), copper,zinc-SOD (Cu,Zn-SOD), catalase (CAT) and glutathione peroxidase (GPx1)], (b) Mn-SOD protein expression, (c) lipid peroxidation markers [malondialdehyde (MDA), 8-epi-prostaglandin-F2α (8-epi-PGF2α)] and by evaluating the possible correlations between tumor biomarkers, reproductive hormone levels and all measured parameters, comprehensively. The data obtained from the patients with EOC (M, n = 26) evaluated according to the histopathological/clinical characteristics of tumors and compared with data of healthy controls [Ctissue (C1) and Cblood/urine (C2), n = 30, respectively). Significantly, low activities of tumor SODtotal (52%), Mn-SOD (42%), Cu,Zn-SOD (55%); high activities of tumor and erythrocyte CAT (66%, 33% respectively) and tumor GPx1 (60%); high levels of tumor Mn-SOD protein expression; tumor MDA (193%) and urinary 8-epi-PGF2α (179%) were observed in serous EOC tumors (M1, n = 18) compared with controls (P < 0.05). However, higher levels of tumor MDA, Mn-SOD protein expression and urinary 8-epi-PGF2α were observed along with lower tumor CAT activity in poorly differentiated or undifferentiated (grade 3, G 3) versus well or moderately well differentiated (grade 1-2, G 1-2) serous EOC tumors. Obtained data indicate the presence of a severe redox imbalance in EOC and draw attention to the criticial role of AOEs in the pathogenesis of the disease. © 2017 IUBMB Life, 69(10):802-813, 2017.


Subject(s)
Carcinoma, Endometrioid/enzymology , Catalase/metabolism , Cystadenocarcinoma, Serous/enzymology , Glutathione Peroxidase/metabolism , Neoplasms, Glandular and Epithelial/enzymology , Ovarian Neoplasms/enzymology , Superoxide Dismutase-1/metabolism , Antioxidants/metabolism , Carcinoma, Endometrioid/diagnosis , Carcinoma, Endometrioid/pathology , Carcinoma, Ovarian Epithelial , Case-Control Studies , Catalase/genetics , Cystadenocarcinoma, Serous/diagnosis , Cystadenocarcinoma, Serous/pathology , Dinoprost/analogs & derivatives , Dinoprost/urine , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Gene Expression , Glutathione Peroxidase/genetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Lipid Peroxidation , Malondialdehyde/blood , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasms, Glandular and Epithelial/diagnosis , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Progesterone/blood , Prolactin/blood , Superoxide Dismutase-1/genetics , Testosterone/blood , Glutathione Peroxidase GPX1
19.
Gynecol Oncol ; 146(1): 170-178, 2017 07.
Article in English | MEDLINE | ID: mdl-28495238

ABSTRACT

OBJECTIVE: Lysyl oxidase (LOX) is an enzyme that catalyzes the cross-linking of collagen and elastin in the extracellular matrix, thus controlling the tensile strength of tissues. Along with this primary function, there are evidences supporting a role for LOX in many critical biological functions, including gene expression regulation, cell growth, adhesion and migration. Accordingly, recent studies have supported a pivotal role for LOX in cancer progression and metastasis. The current study aimed at investigating the prognostic significance and the functional role of intracellular LOX in ovarian cancer. METHODS: To this end, we analyzed LOX expression by immunohistochemistry in archived tumor material from advanced high grade serous ovarian cancer (HGSOC) patients (n=70) and correlated data with clinicopathological parameters and with response to chemotherapy. In vitro experiments were also used to investigate the functional consequences of LOX expression on behavioral aspects of HGSOC cells. RESULTS: Our results showed that nuclear LOX expression is associated with unfavorable outcome in advanced HGSOC, being an independent prognostic factor for disease recurrence. Besides, high nuclear levels were seen to be associated with resistance to first-line chemotherapy. Through gene expression modulation experiments in HGSOC cell lines, we demonstrate that LOX positively regulates cell proliferation, migration and anchorage-independent growth. CONCLUSIONS: Collectively, our data suggest that LOX functions as a tumor promoter in HGSOC and positively regulates several aspects of the metastatic cascade.


Subject(s)
Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/enzymology , Ovarian Neoplasms/enzymology , Protein-Lysine 6-Oxidase/metabolism , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Nucleus/enzymology , Cohort Studies , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Disease Progression , Epithelial-Mesenchymal Transition , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Prognosis , Protein-Lysine 6-Oxidase/biosynthesis , Protein-Lysine 6-Oxidase/genetics
20.
Tohoku J Exp Med ; 241(2): 165-173, 2017 02.
Article in English | MEDLINE | ID: mdl-28202851

ABSTRACT

Ovarian serous carcinoma (OSC) is the most common epithelial ovarian cancer. Inorganic pyrophosphatase (PPA1) catalyzes the hydrolysis of pyrophosphate to inorganic phosphate, thereby providing extra energy for metabolism. The significance of PPA1 in the prognosis of OSC has not been investigated. Our study aimed to explore the expression and predictive role of PPA1 in OSC progression. We screened the expression of PPA1 protein in OSC tissues from 139 patients by immunohistochemistry, and evaluated its correlation with clinicopathological characteristics. PPA1 was categorized as high expression in 58 OSC cases (41.7%), which was correlated with poor differentiation, positive lymph node (LN) metastasis and advanced FIGO (The International Federation of Gynecology and Obstetrics) stages. Univariate and multivariate analyses identified PPA1 as a novel independent prognostic biomarker in OSC patients; meanwhile, conventional factors such as LN status and FIGO stages also showed statistical significance. Moreover, the expression levels of PPA1 protein were higher in A2780 and OVCAR3 human ovarian cancer cell lines than those in normal ovarian surface epithelial cells. Using these ovarian cancer cell lines, we showed that PPA1 overexpression caused the decrease in the expression level of p53, the tumor suppressor, with the increase in ß-catenin level, as determined by Western blot analysis. Conversely, knockdown of PPAI expression was associated with the increase of p53 level and the decreased of ß-catenin level. Consistently, the proliferation and invasion capacities of ovarian cancer cells were enhanced upon PPA1 overexpression. In conclusion, PPA1 serves as a potential prognostic biomarker for patients with OSC.


Subject(s)
Gene Expression Regulation, Neoplastic , Inorganic Pyrophosphatase/genetics , Neoplasms, Glandular and Epithelial/enzymology , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Carcinoma, Ovarian Epithelial , Cell Proliferation , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Female , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Inorganic Pyrophosphatase/metabolism , Kaplan-Meier Estimate , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Prognosis , Tumor Suppressor Protein p53/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...