Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Cancers (Basel) ; 15(13)2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37444425

ABSTRACT

There are strong correlations between the microbiome and human disease, including cancer. However, very little is known about potential mechanisms associated with malignant transformation in microbiome-associated gynecological cancer, except for HPV-induced cervical cancer. Our hypothesis is that differences in bacterial communities in upper genital tract epithelium may lead to selection of specific genomic variation at the cellular level of these tissues that may predispose to their malignant transformation. We first assessed differences in the taxonomic composition of microbial communities and genomic variation between gynecologic cancers and normal samples. Then, we performed a correlation analysis to assess whether differences in microbial communities selected for specific single nucleotide variation (SNV) between normal and gynecological cancers. We validated these results in independent datasets. This is a retrospective nested case-control study that used clinical and genomic information to perform all analyses. Our present study confirms a changing landscape in microbial communities as we progress into the upper genital tract, with more diversity in lower levels of the tract. Some of the different genomic variations between cancer and controls strongly correlated with the changing microbial communities. Pathway analyses including these correlated genes may help understand the basis for how changing bacterial landscapes may lead to these cancers. However, one of the most important implications of our findings is the possibility of cancer prevention in women at risk by detecting altered bacterial communities in the upper genital tract epithelium.

2.
Front Oncol ; 13: 1105504, 2023.
Article in English | MEDLINE | ID: mdl-37287928

ABSTRACT

Objective: To identify high-risk disease in clinicopathologic low-risk endometrial cancer (EC) with high microsatellite instability (MSI-H) or no specific molecular profile (NSMP) and therapeutic insensitivity in clinicopathologic high-risk MSI-H/NSMP EC. Methods: We searched The Cancer Genome Atlas for DNA sequencing, RNA expression, and surveillance data regarding MSI-H/NSMP EC. We used a molecular classification system of E2F1 and CCNA2 expression and sequence variations in POLE, PPP2R1A, or FBXW7 (ECPPF) to prognostically stratify MSI-H/NSMP ECs. Clinical outcomes were annotated after integrating ECPPF and sequence variations in homologous recombination (HR) genes. Results: Data were available for 239 patients with EC, which included 58 MSI-H and 89 NSMP cases. ECPPF effectively stratified MSI-H/NSMP EC into distinct molecular groups with prognostic implications: molecular low risk (MLR), with low CCNA2 and E2F1 expression, and molecular high risk (MHR), with high CCNA2 and E2F1 expression and/or PPP2R1A and/or FBXW7 variants. The 3-year disease-free survival (DFS) rate was 43.8% in the MHR group with clinicopathologic low-risk indicators and 93.9% in the MLR group (P<.001). In the MHR group, wild-type HR genes were present in 28% of cases but in 81% of documented recurrences. The 3-year DFS rate in patients with MSI-H/NSMP EC with clinicopathologic high-risk indicators was significantly higher in the MLR (94.1%) and MHR/HR variant gene (88.9%) groups than in the MHR/HR wild-type gene group (50.3%, P<.001). Conclusion: ECPPF may resolve prognostic challenges for MSI-H/NSMP EC by identifying occult high-risk disease in EC with clinicopathologic low-risk indicators and therapeutic insensitivity in EC with clinicopathologic high-risk indicators.

3.
Clin Transl Med ; 13(5): e1223, 2023 05.
Article in English | MEDLINE | ID: mdl-37165578

ABSTRACT

BACKGROUND: Mutations in the receptor tyrosine kinase gene fibroblast growth factor receptor 2 (FGFR2) occur at a high frequency in endometrial cancer (EC) and have been linked to advanced and recurrent disease. However, little is known about how these mutations drive carcinogenesis. METHODS: Differential transcriptomic analysis and two-step quantitative real-time PCR (qRT-PCR) assays were applied to identify genes differentially expressed in two cohorts of EC patients carrying mutations in the FGFR2 gene as well as in EC cells harbouring mutations in the FGFR2. Candidate genes and target signalling pathways were investigated by qRT-PCR assays, immunohistochemistry and bioinformatics analysis. The functional roles of differently regulated genes were analysed using in vitro and in vivo experiments, including 3D-orthotypic co-culture systems, cell proliferation and migration protocols, as well as colony and focus formation assays together with murine xenograft tumour models. The molecular mechanisms were examined using CRISPR/Cas9-based loss-of-function and pharmacological approaches as well as luciferase reporter techniques, cell-based ectodomain shedding assays and bioinformatics analysis. RESULTS: We show that common FGFR2 mutations significantly enhance the sensitivity to FGF7-mediated activation of a disintegrin and metalloprotease (ADAM)17 and subsequent transactivation of the epidermal growth factor receptor (EGFR). We further show that FGFR2 mutants trigger the activation of ADAM10-mediated Notch signalling in an ADAM17-dependent manner, highlighting for the first time an intimate cooperation between EGFR and Notch pathways in EC. Differential transcriptomic analysis in EC cells in a cohort of patients carrying mutations in the FGFR2 gene identified a strong association between FGFR2 mutations and increased expression of members of the Notch pathway and ErbB receptor family. Notably, FGFR2 mutants are not constitutively active but require FGF7 stimulation to reprogram Notch and EGFR pathway components, resulting in ADAM17-dependent oncogenic growth. CONCLUSIONS: These findings highlight a pivotal role of ADAM17 in the pathogenesis of EC and provide a compelling rationale for targeting ADAM17 protease activity in FGFR2-driven cancers.


Subject(s)
Endometrial Neoplasms , Receptor, Fibroblast Growth Factor, Type 2 , Female , Humans , Mice , Animals , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Signal Transduction/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Mutation/genetics
4.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555654

ABSTRACT

Endometrial cancer (EC) incidence and mortality continues to rise. Molecular profiling of EC promises improvement of risk assessment and treatment selection. However, we still lack robust and accurate models to predict those at risk of failing treatment. The objective of this pilot study is to create models with clinical and genomic data that will discriminate patients with EC at risk of disease recurrence. We performed a pilot, retrospective, case−control study evaluating patients with EC, endometrioid type: 7 with recurrence of disease (cases), and 55 without (controls). RNA was extracted from frozen specimens and sequenced (RNAseq). Genomic features from RNAseq included transcriptome expression, genomic, and structural variation. Feature selection for variable reduction was performed with univariate ANOVA with cross-validation. Selected variables, informative for EC recurrence, were introduced in multivariate lasso regression models. Validation of models was performed in machine-learning platforms (ML) and independent datasets (TCGA). The best performing prediction models (out of >170) contained the same lncRNA features (AUC of 0.9, and 95% CI: 0.75, 1.0). Models were validated with excellent performance in ML platforms and good performance in an independent dataset. Prediction models of EC recurrence containing lncRNA features have better performance than models with clinical data alone.


Subject(s)
Carcinoma, Endometrioid , Endometrial Neoplasms , RNA, Long Noncoding , Female , Humans , Retrospective Studies , Case-Control Studies , Pilot Projects , Neoplasm Recurrence, Local/genetics , Carcinoma, Endometrioid/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/epidemiology , Genomics
5.
Int J Mol Sci ; 23(23)2022 Nov 26.
Article in English | MEDLINE | ID: mdl-36499142

ABSTRACT

The preoperative diagnosis of pelvic masses has been elusive to date. Methods for characterization such as CA-125 have had limited specificity. We hypothesize that genomic variation can be used to create prediction models which accurately distinguish high grade serous ovarian cancer (HGSC) from benign tissue. METHODS: In this retrospective, pilot study, we extracted DNA and RNA from HGSC specimens and from benign fallopian tubes. Then, we performed whole exome sequencing and RNA sequencing, and identified single nucleotide variants (SNV), copy number variants (CNV) and structural variants (SV). We used these variants to create prediction models to distinguish cancer from benign tissue. The models were then validated in independent datasets and with a machine learning platform. RESULTS: The prediction model with SNV had an AUC of 1.00 (95% CI 1.00-1.00). The models with CNV and SV had AUC of 0.87 and 0.73, respectively. Validated models also had excellent performances. CONCLUSIONS: Genomic variation of HGSC can be used to create prediction models which accurately discriminate cancer from benign tissue. Further refining of these models (early-stage samples, other tumor types) has the potential to lead to detection of ovarian cancer in blood with cell free DNA, even in early stage.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Female , Humans , Cystadenocarcinoma, Serous/diagnosis , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Fallopian Tube Neoplasms/pathology , Fallopian Tubes/pathology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Pilot Projects , Retrospective Studies , Genome
6.
PLoS One ; 17(12): e0278408, 2022.
Article in English | MEDLINE | ID: mdl-36454788

ABSTRACT

In endometrial cancer, occult high-risk subtypes (rooted in histomorphologically low-risk disease) with insensitivity to adjuvant therapies impede improvements in therapeutic efficacy. Therefore, we aimed to assess the ability of molecular high-risk (MHR) and low-risk (MLR) ECPPF (E2F1, CCNA2, POLE, PPP2R1A, FBXW7) stratification to profile recurrence in early, low-risk endometrioid endometrial cancer (EEC) and insensitivity to platinum-based chemotherapy or radiotherapy (or both) in high-risk EEC. Using The Cancer Genome Atlas endometrial cancer database, we identified 192 EEC cases with available DNA sequencing and RNA expression data. Molecular parameters were integrated with clinicopathologic risk factors and adverse surveillance events. MHR was defined as high (-H) CCNA2 or E2F1 log2 expression (≥2.75), PPP2R1A mutations (-mu), or FBXW7mu; MLR was defined as low (-L) CCNA2 and E2F1 log2 expression (<2.75). We assessed 164 cases, plus another 28 with POLEmu for favorable-outcomes comparisons. MHR and MLR had significantly different progression-free survival (PFS) rates (P < .001), independent of traditional risk factors (eg, TP53mu), except for stage IV disease. PFS of CCNA2-L/E2F1-L paralleled that of POLEmu. ECPPF status stratified responses to adjuvant therapy in stage III-IV EEC (P < .01) and profiled stage I, grade 1-2 cases with risk of recurrence (P < .001). MHR was associated with CTNNB1mu-linked treatment failures (P < .001). Expression of homologous recombination repair (HR) and cell cycle genes was significantly elevated in CCNA2-H/E2F1-H compared with CCNA2-L/E2F1-L (P<1.0E-10), suggesting that HR deficiencies may underlie the favorable PFS in MLR. HRmu were detected in 20.7%. No treatment failures were observed in high-grade or advanced EEC with HRmu (P = .02). Favorable PFS in clinically high-risk EEC was associated with HRmu and MLR ECPPF (P < .001). In summary, MLR ECPPF and HRmu were associated with therapeutic efficacy in EEC. MHR ECPPF was associated with low-risk, early-stage recurrences and insensitivity to adjuvant therapies.


Subject(s)
Carcinoma, Endometrioid , Endometrial Neoplasms , Female , Humans , Genes, cdc , F-Box-WD Repeat-Containing Protein 7/genetics , Genes, Regulator , Transcription Factors , Endometrial Neoplasms/genetics , Endometrial Neoplasms/therapy , E2F1 Transcription Factor , Cyclin A2 , Protein Phosphatase 2/genetics
7.
Womens Health Rep (New Rochelle) ; 3(1): 678-685, 2022.
Article in English | MEDLINE | ID: mdl-36147832

ABSTRACT

Objectives: Rural ovarian cancer patients experience worse survival compared to urban patients. We assessed whether distance to gynecologic oncology specialists was associated with survival for patients in a rural state. Methods: Demographic, tumor, and treatment characteristics were extracted from the Iowa Cancer Registry for patients diagnosed between 1990 and 2018. Data were linked to the county-level 2018-2019 Area Health Resource File (number of surgeons and hospital beds per 100,000 population). Rurality was defined using 2013 Rural-Urban Continuum Codes; distance to the nearest gynecologic oncologist was calculated from the centroid of the county of residence to the centroid of the nearest county with a high volume health care center with a gynecologic oncologist. Associations with survival were assessed using multivariable Cox proportional hazards models. Results: Analyses included 1,562 ovarian cancer patients. Mean distance to gynecologic oncology was 60.8 miles, and median survival was 23 months. Unadjusted models showed increased distance from gynecologic oncology had progressively greater risk of death 30-49 miles (hazard ratio [HR] = 1.09, confidence interval [CI]: 1.04-1.15), 50-69 miles (HR = 1.19, CI: 1.07-1.32), 70+ miles (HR = 1.30, CI: 1.11-1.51). In adjusted models, association of distance to gynecologic oncology with risk of death was not significant; however, more advanced cancer stage and age, unmarried status, and higher county-level poverty were independently associated with increased risk of death. Conclusions: Above and beyond demographics and stage, distance to gynecologic oncology care was not an independent predictor of ovarian cancer survival. Further studies are needed to determine how to mitigate the factors contributing to worsened ovarian cancer survival among rural patients.

8.
Cancers (Basel) ; 14(14)2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35884615

ABSTRACT

Advanced high-grade serous (HGSC) ovarian cancer is treated with either primary surgery followed by chemotherapy or neoadjuvant chemotherapy followed by interval surgery. The decision to proceed with surgery primarily or after chemotherapy is based on a surgeon's clinical assessment and prediction of an optimal outcome. Optimal and complete cytoreductive surgery are correlated with improved overall survival. This clinical assessment results in an optimal surgery approximately 70% of the time. We hypothesize that this prediction can be improved by using biological tumor data to predict optimal cytoreduction. With access to a large biobank of ovarian cancer tumors, we obtained genomic data on 83 patients encompassing gene expression, exon expression, long non-coding RNA, micro RNA, single nucleotide variants, copy number variation, DNA methylation, and fusion transcripts. We then used statistical learning methods (lasso regression) to integrate these data with pre-operative clinical information to create predictive models to discriminate which patient would have an optimal or complete cytoreductive outcome. These models were then validated within The Cancer Genome Atlas (TCGA) HGSC database and using machine learning methods (TensorFlow). Of the 124 models created and validated for optimal cytoreduction, 21 performed at least equal to, if not better than, our historical clinical rate of optimal debulking in advanced-stage HGSC as a control. Of the 89 models created to predict complete cytoreduction, 37 have the potential to outperform clinical decision-making. Prospective validation of these models could result in improving our ability to objectively predict which patients will undergo optimal cytoreduction and, therefore, improve our ovarian cancer outcomes.

9.
Acta Obstet Gynecol Scand ; 101(10): 1085-1092, 2022 10.
Article in English | MEDLINE | ID: mdl-35778930

ABSTRACT

INTRODUCTION: The survival benefits of surgical cytoreduction in ovarian cancer are well-established. However, the surgical outcome has never been assessed while controlling for the efficacy of chemotherapy. This leaves the possibility that cytoreduction may not be beneficial for patients whose cancer does not respond well to adjuvant treatment. We sought to answer whether surgical cytoreduction independently improves overall survival when controlling for chemotherapy outcome. MATERIAL AND METHODS: We performed a retrospective case-control study using our institution's ovarian cancer database to evaluate the effect of optimal cytoreduction on advanced stage, high-grade serous ovarian cancer. Patients' characteristics were compared using both univariate and multivariate regression modeling to assess for independent predictors of overall survival. RESULTS: A total of 470 patients were assessed for inclusion; 234 responders to chemotherapy and 98 nonresponders. Significant survival characteristics were identified and included in the multivariate analysis. Independent predictors of survival in the multivariate analysis were age, responder status, optimal cytoreduction, neoadjuvant chemotherapy, and number of chemotherapy cycles. Kaplan-Meier survival curves showed improved survival for both patients who responded to chemotherapy and for those undergoing optimal cytoreduction (p < 0.001). We also demonstrated improved survival for patients receiving optimal cytoreduction among both nonresponders and responders (p < 0.001). CONCLUSIONS: Our analysis shows that patients who undergo optimal cytoreduction have an overall survival benefit regardless of their response to chemotherapy. Therefore, cytoreduction should be considered in all patients, even in those with advanced disease, if an optimal result can be achieved. This study was underpowered to assess patients who received neoadjuvant chemotherapy as a separate subgroup, but the order of treatment was controlled for in the overall analysis.


Subject(s)
Cytoreduction Surgical Procedures , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial/surgery , Case-Control Studies , Chemotherapy, Adjuvant , Female , Humans , Neoplasm Staging , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Retrospective Studies
10.
Int J Gynecol Cancer ; 32(4): 540-546, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35197327

ABSTRACT

OBJECTIVE: To determine if there is a difference in overall survival of patients with epithelial ovarian cancer in rural, urban, and metropolitan settings in the United States. METHODS: We performed a retrospective cohort study using 2004-2016 National Cancer Database (NCDB) data including high and low grade, stage I-IV disease. Bivariate analyses used Student's t-test for continuous variables and χ2 test for dichotomous variables. Kaplan-Meier curves estimated survival of patients based on location of residence, and univariate analyses using Cox proportional HR assessed survival based on baseline characteristics. Multivariate analysis was performed to account for significant covariates. Propensity score matching was used to validate the multivariate survival model. For all tests, p<0.05 was considered statistically significant. RESULTS: A total of 111 627 patients were included with a mean age of 62.5 years for metroolitan (range 18-90), 64.0 years for rural (range 19-90) and 63.2 years for urban areas (range 18-90). Of all patients included, 94 290 were in a metropolitan area (counties >1 million population or 50 000-999 999), 15 386 were in an urban area (population of 10 000-49 999), and 1951 were in a rural area (non-metropolitan/non-core population). Univariate Cox proportional hazards models showed clinically significant differences in survival in patients from metropolitan, urban, and rural areas. Multivariate Cox proportional hazards models showed a clinically significant increase in HRs for patients in rural settings (HR 1.17; 95% CI 1.06 to 1.29). Increasing age and stage, non-insured status, non-white race, and comorbidity were also significant for poorer survival. CONCLUSION: Patients with ovarian cancer who live in rural settings with small populations and greater distance to tertiary care centers have poorer survival. These differences hold after controlling for stage, age, and other significant risk factors related to poorer outcomes. To improve clinical outcomes, we need further studies to identify which of these factors are actionable.


Subject(s)
Ovarian Neoplasms , Rural Population , Carcinoma, Ovarian Epithelial , Humans , Middle Aged , Proportional Hazards Models , Retrospective Studies , United States/epidemiology , Urban Population
11.
Pharmaceuticals (Basel) ; 14(9)2021 Sep 21.
Article in English | MEDLINE | ID: mdl-34577642

ABSTRACT

Elevated expression of placenta-specific protein 1 (PLAC1) is associated with the increased proliferation and invasiveness of a variety of human cancers, including ovarian cancer. Recent studies have shown that the tumor suppressor p53 directly suppresses PLAC1 transcription. However, mutations in p53 lead to the loss of PLAC1 transcriptional suppression. Small molecules that structurally convert mutant p53 proteins to wild-type conformations are emerging. Our objective was to determine whether the restoration of the wild-type function of mutated p53 could rescue PLAC1 transcriptional suppression in tumors harboring certain TP53 mutations. Ovarian cancer cells OVCAR3 and ES-2, both harboring TP53 missense mutations, were treated with the p53 reactivator HO-3867. Treatment with HO-3867 successfully rescued PLAC1 transcriptional suppression. In addition, cell proliferation was inhibited and cell death through apoptosis was increased in both cell lines. We conclude that the use of HO-3867 as an adjuvant to conventional therapeutics in ovarian cancers harboring TP53 missense mutations could improve patient outcomes. Validation of this conclusion must, however, come from an appropriately designed clinical trial.

12.
Oncol Lett ; 22(4): 733, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34429773

ABSTRACT

The 8q24 chromosomal region is strongly associated with an increased risk of ovarian cancer. One single nucleotide polymorphism that is associated with ovarian cancer in this region is rs6983267, located within the long non-coding RNA colon cancer associated transcript 2 (CCAT2). The aim of the present study was to assess the association between rs6983267 and clinical outcomes in patients with high-grade serous ovarian cancer (HGSOC). The present retrospective genetic association study utilized Sanger sequencing to determine the genotype at the rs6983267 locus (GG, GT, TT) in 98 patients with HGSOC. Survival time and chemotherapy responses between patients were compared with the TT genotype and patients with a genotype containing a G allele (GT, GG). Survival analyses were performed using Cox proportional hazard ratio analysis. Association with chemo-response was performed using a logistic regression. The results revealed that patients with HGSOC and the TT genotype at the rs6983267 locus had improved survival time compared with patients with genotypes containing a G allele [hazard ratio=0.59; 95% confidence interval (CI), 0.36-0.97; P=0.039] and were significantly associated with International Federation of Gynecology and Obstetrics stage [odds ratio (OR)=5.34; 95% CI, 1.50-22.62; P=0.014] and positive chemo-response (OR=4.51; 95% CI, 1.40-18.00; P=0.018). In summary, patients with HGSOC and the TT genotype at the rs6983267 locus had improved survival time compared with those with a G allele, despite being associated with more advanced disease; this was possibly due to an improved response to chemotherapy.

13.
Am J Reprod Immunol ; 86(4): e13469, 2021 10.
Article in English | MEDLINE | ID: mdl-34022086

ABSTRACT

OBJECTIVE: Treatment of high-grade serous ovarian cancer (HGSOC) will benefit from early detection of cancer. Here, we provide proof-of-concept data supporting the hypothesis that circulating immune cells, because of their early recognition of tumors and the tumor microenvironment, can be considered for biomarker discovery. METHODS: Longitudinal blood samples from C57BL/6 mice bearing syngeneic ovarian tumors and peripheral blood mononuclear cells (PBMC) from healthy postmenopausal women and newly diagnosed for HGSOC patients were subjected to RNASeq. The results from human immune cells were validated using Affymetrix microarrays. Differentially expressed transcripts in immune cells from tumor-bearing mice and HGSOC patients were compared to matching controls. RESULTS: A total of 1282 transcripts (798 and 484, up- and downregulated, respectively) were differentially expressed in the tumor-bearing mice as compared with controls. Top 100 genes showing longitudinal changes in gene expression 2, 4, 7, and 18 days after tumor implantation were identified. Analysis of the PBMC from healthy post-menopausal women and HGSOC patients identified 4382 differentially expressed genes and 519 of these were validated through Affymetrix microarray analysis. A total of 384 genes, including IL-1R2, CH3L1, Infitm1, FP42, CXC42, Hdc, Spib, and Sema6b, were differentially expressed in the human and mouse datasets. CONCLUSION: The PBMC transcriptome shows longitudinal changes in response to the progressing tumor. Several potential biomarker transcripts were identified in HGSOC patients and mouse models. Monitoring their expression in individual PBMC subsets can serve as additional discriminator for the diagnosis of HGSOC.


Subject(s)
Cystadenocarcinoma, Serous/diagnosis , Ovarian Neoplasms/diagnosis , Tumor Microenvironment , Animals , Biomarkers, Tumor , Cell Line , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Female , Gene Expression Profiling , Humans , Leukocytes, Mononuclear/metabolism , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proof of Concept Study , Transcriptome
14.
Cancers (Basel) ; 13(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807612

ABSTRACT

Bacteria, archaea, and viruses are associated with numerous human cancers. To date, microbiome variations in transcription have not been evaluated relative to upper female genital tract cancer risk. Our aim was to assess differences in bacterial, archaea, and viral transcript (BAVT) expression between different gynecological cancers and normal fallopian tubes. In this case-control study we performed RNA sequencing on 12 normal tubes, 112 serous ovarian cancers (HGSC) and 62 endometrioid endometrial cancers (EEC). We used the centrifuge algorithm to classify resultant transcripts into four indexes: bacterial, archaea, viral, and human genomes. We then compared BAVT expression from normal samples, HGSC and EEC. T-test was used for univariate comparisons (correcting for multiple comparison) and lasso for multivariate modelling. For validation we performed DNA sequencing of normal tubes in comparison to HGSC and EEC BAVTs in the TCGA database. Pathway analyses were carried out to evaluate the function of significant BAVTs. Our results show that BAVT expression levels vary between different gynecological cancers. Finally, we mapped some of these BAVTs to the human genome. Numerous map locations were close to regulatory genes and long non-coding RNAs based on the pathway enrichment analysis. BAVTs may affect gynecological cancer risk and may be part of potential targets for cancer therapy.

15.
Gynecol Oncol ; 162(1): 182-189, 2021 07.
Article in English | MEDLINE | ID: mdl-33867147

ABSTRACT

OBJECTIVE: PI3K-AKT pathway mutations initiate a kinase cascade that characterizes endometrial cancer (EC). As kinases seldom cause oncogenic transformation without dysregulation of antagonistic phosphatases, pivotal interactions governing this pathway were explored and correlated with clinical outcomes. METHODS: After exclusion of patients with POLE mutations from The Cancer Genome Atlas EC cohort with endometrioid or serous EC, the study population was 209 patients with DNA sequencing, quantitative gene-specific RNA expression, copy number variation (CNV), and surveillance data available. Extracted data were annotated and integrated. RESULTS: A PIK3CA, PTEN, or PIK3R1 mutant (-mu) was present in 83% of patients; 57% harbored more than 1 mutation without adversely impacting progression-free survival (PFS) (P = .10). PIK3CA CNV of at least 1.1 (CNV high [-H]) was detected in 26% and linked to TP53-mu and CIP2A expression (P < .001) but was not associated with PFS (P = .24). PIK3CA expression was significantly different between those with CIP2A-H and CIP2A low (-L) expression (the endogenous inhibitor of protein phosphatase 2A [PP2A]), when stratified by PIK3CA mutational status or by PIK3CA CNV-H and CNV-L (all P < .01). CIP2A-H or PPP2R1A-mu mitigates PP2A kinase dephosphorylation, and FBXW7-mu nullifies E3 ubiquitin ligase (E3UL) oncoprotein degradation. CIP2A-H and PPP2R1A-mu (PP2A impairment) and FBXW7-mu (E3UL impairment) were associated with compromised PFS (P < .001) and were prognostically discriminatory for PIK3CA-mu and PIK3CA CNV-H tumors (P < .001). Among documented recurrences, 84% were associated with impaired PP2A (75%) and/or E3UL (20%). CONCLUSION: PP2A and E3UL deficiencies are seminal biological drivers in EC independent of PIK3CA-mu, PTEN-mu, and PIK3R1-mu and PIK3CA CNV.


Subject(s)
Endometrial Neoplasms/enzymology , Protein Phosphatase 2/deficiency , Ubiquitin-Protein Ligases/deficiency , Abdominal Neoplasms , Autoantigens/biosynthesis , Autoantigens/genetics , Class I Phosphatidylinositol 3-Kinases/biosynthesis , Class I Phosphatidylinositol 3-Kinases/genetics , Class Ia Phosphatidylinositol 3-Kinase/biosynthesis , Class Ia Phosphatidylinositol 3-Kinase/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Female , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mutation , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
16.
Sci Rep ; 11(1): 5957, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33727600

ABSTRACT

Nearly a third of patients with high-grade serous ovarian cancer (HGSC) do not respond to initial therapy and have an overall poor prognosis. However, there are no validated tools that accurately predict which patients will not respond. Our objective is to create and validate accurate models of prediction for treatment response in HGSC. This is a retrospective case-control study that integrates comprehensive clinical and genomic data from 88 patients with HGSC from a single institution. Responders were those patients with a progression-free survival of at least 6 months after treatment. Only patients with complete clinical information and frozen specimen at surgery were included. Gene, miRNA, exon, and long non-coding RNA (lncRNA) expression, gene copy number, genomic variation, and fusion-gene determination were extracted from RNA-sequencing data. DNA methylation analysis was performed. Initial selection of informative variables was performed with univariate ANOVA with cross-validation. Significant variables (p < 0.05) were included in multivariate lasso regression prediction models. Initial models included only one variable. Variables were then combined to create complex models. Model performance was measured with area under the curve (AUC). Validation of all models was performed using TCGA HGSC database. By integrating clinical and genomic variables, we achieved prediction performances of over 95% in AUC. Most performances in the validation set did not differ from the training set. Models with DNA methylation or lncRNA underperformed in the validation set. Integrating comprehensive clinical and genomic data from patients with HGSC results in accurate and robust prediction models of treatment response.


Subject(s)
Biomarkers, Tumor , Cystadenocarcinoma, Serous/diagnosis , Disease Susceptibility , Models, Biological , Ovarian Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Case-Control Studies , Combined Modality Therapy , Computational Biology/methods , Cystadenocarcinoma, Serous/etiology , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/therapy , DNA Methylation , Disease Management , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genomics/methods , Humans , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasm, Residual/diagnosis , Ovarian Neoplasms/etiology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Prognosis , Reproducibility of Results , Retrospective Studies , Treatment Outcome
17.
Int J Mol Sci ; 22(3)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499129

ABSTRACT

Long non-coding RNA's (lncRNA) are RNA sequences that do not encode proteins and are greater than 200 nucleotides in length. They regulate complex cellular mechanisms and have been associated with prognosis in various types of cancer. We aimed to identify lncRNA sequences that are associated with high grade serous ovarian cancer (HGSC) and assess their impact on overall survival. RNA was extracted from 112 HGSC patients and 12 normal fallopian tube samples from our Biobank tissue repository. RNA was sequenced and the Ultrafast and Comprehensive lncRNA detection and quantification pipeline (UClncR) was used for the identification of lncRNA sequences. Univariate logistic and multivariate lasso regression analyses identified lncRNA that was associated with HGSC. Univariate and multivariate Cox proportional hazard ratios were used to evaluate independent predictors of survival. 1943 of 16,325 investigated lncRNA's were differentially expressed in HGSC as compared to controls (p < 0.001). Nine of these demonstrated association with cancer after multivariate lasso regression. Our multivariate analysis of survival identified four lncRNA's associated with survival in HGSC. Three out of these four were found to be independently significant after accounting for all clinical covariates. Lastly, seven lncRNAs were independently associated with initial response to chemotherapy; four portended a worse response, while three were associated with improved response. More research is needed, but there is potential for these lncRNAs to be used as biomarkers of HGSC or predictors of treatment outcome in the future.


Subject(s)
Ovarian Neoplasms/genetics , RNA, Long Noncoding/genetics , Antineoplastic Agents/pharmacology , Biological Specimen Banks , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/mortality , Case-Control Studies , Fallopian Tubes/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome, Human , Genomics , Humans , Kaplan-Meier Estimate , Multivariate Analysis , Ovarian Neoplasms/mortality , Proportional Hazards Models , RNA-Seq , Retrospective Studies , Time Factors , Treatment Outcome
18.
Int J Oncol ; 57(6): 1348-1357, 2020 12.
Article in English | MEDLINE | ID: mdl-33174010

ABSTRACT

Recently, the compilation of massive amounts of genetic and genomic information on a wide variety of human cancer types, collectively known as The Cancer Genome Atlas (TCGA), has revealed a wealth of descriptive classification schemes both within and between different types and sources of cancer. In endometrial cancer, TCGA analyses have produced a post hoc scheme composed of four clusters: DNA polymerase ε catalytic subunit A (POLE) ultra­mutated (cluster 1), microsatellite instability (MSI) hypermutated (cluster 2), copy­number low (endometrioid, cluster 3) and copy­number high (serous­like, cluster 4). Given that cultured cells are the pre­clinical platform of cancer research, it was questioned how representative endometrial cancer cultured cell lines are in the context of TCGA­driven classification scheme. To address this issue in endometrial cancer cell lines, the present study investigated five commonly used cell lines: Ishikawa, ECC­1, Hec50co, KLE And RL95­2. The histology, mutation profile, MutL homolog 1 promoter methylation, copy­number variation, homologous recombination repair and microsatellite instability in each of these cell lines was assessed. The result of this characterization was that none of the cell lines fits neatly into any one of TCGA classes but are still useful models for groups of endometrial tumors. Furthermore, the contention that the ECC­1 cell line is actually Ishikawa was addressed using additional data. It was confirmed that ECC­1 cells likely no longer exist as ECC­1 but that they are not exactly Ishikawa either. For this reason, ECC­1 cells are suggested to be used in vitro but with this caveat in mind. Finally, we compiled a database of 127 endometrial cancer cell lines, including the five reported on here. The wide range of variation found in these cell lines highlights the need to further characterize these cells to select models that are more representative of the various histological and genomic aspects of endometrial cancer.


Subject(s)
Endometrial Neoplasms/genetics , Endometrium/pathology , Cell Line, Tumor , DNA Copy Number Variations , DNA Polymerase II/genetics , Endometrial Neoplasms/classification , Endometrial Neoplasms/pathology , Female , Humans , Microsatellite Instability , Mutation , Poly-ADP-Ribose Binding Proteins/genetics , Recombinational DNA Repair
19.
Int J Gynecol Cancer ; 30(6): 845-852, 2020 06.
Article in English | MEDLINE | ID: mdl-32341114

ABSTRACT

OBJECTIVE: In previous studies, neoadjuvant chemotherapy followed by interval debulking surgery was not inferior to primary cytoreductive surgery as initial treatment for advanced epithelial ovarian cancer. Our study aimed to compare surgical and survival outcomes between the two treatments in a large national database. METHODS: Data were extracted from the National Cancer Database from January 2004 to December 2015. Patients with FIGO (International Federation of Gynecologists and Obstetricians) stage III-IV epithelial ovarian cancer and known sequence of treatment were included: primary cytoreductive (surgery=26 717 and neoadjuvant chemotherapy=9885). Tubal and primary peritoneal cancer diagnostic codes were not included. Residual disease after treatment was defined based on recorded data: R0 defined as microscopic or no residual disease; R1 defined as macroscopic residual disease. Multivariate Cox proportional HR was used for survival analysis. Multivariate logistic regression analysis was utilized to compare mortality between groups. Outcomes were adjusted for significant covariates. Validation was performed using propensity score matching of significant covariates. RESULTS: A total of 36 602 patients were included in the analysis. Patients who underwent primary cytoreductive surgery had better survival than those treated with neoadjuvant chemotherapy followed by interval surgery, after adjusting for age, co-morbidities, stage, and residual disease (p<0.001). Primary cytoreductive surgery patients with R0 disease had best median survival (62.6 months, 95% CI 60.5-64.5). Neoadjuvant chemotherapy patients with R1 disease had worst median survival (29.5 months, 95% CI 28.4-31.9). There were small survival differences between primary cytoreductive surgery with R1 (38.9 months) and neoadjuvant chemotherapy with R0 (41.8 months) (HR 0.93, 95% CI 0.87 to 1.0), after adjusting for age, co-morbidities, grade, histology, and stage. Neoadjuvant chemotherapy had 3.5 times higher 30-day mortality after surgery than primary cytoreductive surgery (95% CI 2.46 to 5.64). The 90-day mortality was higher for neoadjuvant chemotherapy in multivariate analysis (HR 1.31, 95% CI 1.06 to 1.61) but similar to primary cytoreductive surgery after excluding high-risk patients. CONCLUSIONS: Most patients with advanced epithelial ovarian cancer may benefit from primary cytoreductive surgery. Patients treated with neoadjuvant chemotherapy should be those with co-morbidities unfit for surgery.


Subject(s)
Carcinoma, Ovarian Epithelial/surgery , Cytoreduction Surgical Procedures , Neoadjuvant Therapy , Ovarian Neoplasms/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/mortality , Female , Humans , Middle Aged , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Retrospective Studies , Survival Analysis , United States/epidemiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...