Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
2.
Gynecol Oncol ; 178: 119-129, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37862791

ABSTRACT

OBJECTIVE: This prespecified exploratory analysis evaluated the association of gene expression signatures, tumor mutational burden (TMB), and multiplex immunohistochemistry (mIHC) tumor microenvironment-associated cell phenotypes with clinical outcomes of pembrolizumab in advanced recurrent ovarian cancer (ROC) from the phase II KEYNOTE-100 study. METHODS: Pembrolizumab-treated patients with evaluable RNA-sequencing (n = 317), whole exome sequencing (n = 293), or select mIHC (n = 125) data were evaluated. The association between outcomes (objective response rate [ORR], progression-free survival [PFS], and overall survival [OS]) and gene expression signatures (T-cell-inflamed gene expression profile [TcellinfGEP] and 10 non-TcellinfGEP signatures), TMB, and prespecified mIHC cell phenotype densities as continuous variables was evaluated using logistic (ORR) and Cox proportional hazards regression (PFS; OS). One-sided p-values were calculated at prespecified α = 0.05 for TcellinfGEP, TMB, and mIHC cell phenotypes and at α = 0.10 for non-TcellinfGEP signatures; all but TcellinfGEP and TMB were adjusted for multiplicity. RESULTS: No evidence of associations between ORR and key axes of gene expression was observed. Negative associations were observed between outcomes and TcellinfGEP-adjusted glycolysis (PFS, adjusted-p = 0.019; OS, adjusted-p = 0.085) and hypoxia (PFS, adjusted-p = 0.064) signatures. TMB as a continuous variable was not associated with outcomes (p > 0.05). Positive associations were observed between densities of myeloid cell phenotypes CD11c+ and CD11c+/MHCII-/CD163-/CD68- in the tumor compartment and ORR (adjusted-p = 0.025 and 0.013, respectively). CONCLUSIONS: This exploratory analysis in advanced ROC did not find evidence for associations between gene expression signatures and outcomes of pembrolizumab. mIHC analysis suggests CD11c+ and CD11c+/MHCII-/CD163-/CD68- phenotypes representing myeloid cell populations may be associated with improved outcomes with pembrolizumab in advanced ROC. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov, NCT02674061.


Subject(s)
Antineoplastic Agents, Immunological , Ovarian Neoplasms , Humans , Female , Antineoplastic Agents, Immunological/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Progression-Free Survival , Carcinoma, Ovarian Epithelial/drug therapy , Biomarkers, Tumor/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/chemically induced , Tumor Microenvironment
3.
Sci Rep ; 13(1): 3334, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36849518

ABSTRACT

Epithelial ovarian cancer is the most lethal gynecological malignancy, owing notably to its high rate of therapy-resistant recurrence in spite of good initial response to chemotherapy. Although poly(ADP-ribose) polymerase inhibitors (PARPi) have shown promise for ovarian cancer treatment, extended therapy usually leads to acquired PARPi resistance. Here we explored a novel therapeutic option to counter this phenomenon, combining PARPi and inhibitors of nicotinamide phosphoribosyltransferase (NAMPT). Cell-based models of acquired PARPi resistance were created through an in vitro selection procedure. Using resistant cells, xenograft tumors were grown in immunodeficient mice, while organoid models were generated from primary patient tumor samples. Intrinsically PARPi-resistant cell lines were also selected for analysis. Our results show that treatment with NAMPT inhibitors effectively sensitized all in vitro models to PARPi. Adding nicotinamide mononucleotide, the resulting NAMPT metabolite, abrogated the therapy-induced cell growth inhibition, demonstrating the specificity of the synergy. Treatment with olaparib (PARPi) and daporinad (NAMPT inhibitor) depleted intracellular NAD+ , induced double-strand DNA breaks, and promoted apoptosis as monitored by caspase-3 cleavage. The two drugs were also synergistic in mouse xenograft models and clinically relevant patient-derived organoids. Therefore, in the context of PARPi resistance, NAMPT inhibition could offer a promising new option for ovarian cancer patients.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Humans , Animals , Mice , Female , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Niacinamide , Ovarian Neoplasms/drug therapy , Dinucleoside Phosphates
4.
Sci Rep ; 11(1): 18183, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521878

ABSTRACT

Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer. Among the key challenges in developing effective therapeutics is the poor translation of preclinical models used in the drug discovery pipeline. This leaves drug attrition rates and costs at an unacceptably high level. Previous work has highlighted the discrepancies in therapeutic response between current in vitro and in vivo models. To address this, we conducted a comparison study to differentiate the carboplatin chemotherapy response across four different model systems including 2D monolayers, 3D spheroids, 3D ex vivo tumors and mouse xenograft models. We used six previously characterized EOC cell lines of varying chemosensitivity and performed viability assays for each model. In vivo results from the mouse model correlated with 2D response in 3/6 cell lines while they correlated with 3D spheroids and the ex vivo model in 4/6 and 5/5 cell lines, respectively. Our results emphasize the variability in therapeutic response across models and demonstrate that the carboplatin response in EOC cell lines cultured in a 3D ex vivo model correlates best with the in vivo response. These results highlight a more feasible, reliable, and cost-effective preclinical model with the highest translational potential for drug screening and prediction studies in EOC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carboplatin/therapeutic use , Drug Resistance, Neoplasm , Ovarian Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/pharmacology , Carboplatin/pharmacology , Cell Line, Tumor , Female , Humans , Mice , Spheroids, Cellular/drug effects , Tumor Cells, Cultured
5.
Int J Mol Sci ; 22(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070214

ABSTRACT

During tubo-ovarian high-grade serous carcinoma (HGSC) progression, tumoral cells undergo phenotypic changes in their epithelial marker profiles, which are essential for dissemination processes. Here, we set out to determine whether standard epithelial markers can predict HGSC patient prognosis. Levels of E-CADH, KRT7, KRT18, KRT19 were quantified in 18 HGSC cell lines by Western blot and in a Discovery cohort tissue microarray (TMA) (n = 101 patients) using immunofluorescence. E-CADH and KRT7 levels were subsequently analyzed in the TMA of the Canadian Ovarian Experimental Unified Resource cohort (COEUR, n = 1158 patients) and in public datasets. Epithelial marker expression was highly variable in HGSC cell lines and tissues. In the Discovery cohort, high levels of KRT7 and KRT19 were associated with an unfavorable prognosis, whereas high E-CADH expression indicated a better outcome. Expression of KRT7 and E-CADH gave a robust combination to predict overall survival (OS, p = 0.004) and progression free survival (PFS, p = 5.5 × 10-4) by Kaplan-Meier analysis. In the COEUR cohort, the E-CADH-KRT7 signature was a strong independent prognostic biomarker (OS, HR = 1.6, p = 2.9 × 10-4; PFS, HR = 1.3, p = 0.008) and predicted a poor patient response to chemotherapy (p = 1.3 × 10-4). Our results identify a combination of two epithelial markers as highly significant indicators of HGSC patient prognosis and treatment response.


Subject(s)
Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Cadherins/metabolism , Cystadenocarcinoma, Serous/metabolism , Fallopian Tube Neoplasms/metabolism , Keratin-7/metabolism , Ovarian Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cohort Studies , Cystadenocarcinoma, Serous/mortality , Fallopian Tube Neoplasms/mortality , Female , Humans , Kaplan-Meier Estimate , Keratin-19/metabolism , Lung Neoplasms/metabolism , Middle Aged , Ovarian Neoplasms/mortality , Prognosis , Progression-Free Survival , Stomach Neoplasms/metabolism , Vimentin/metabolism
6.
Mod Pathol ; 34(1): 194-206, 2021 01.
Article in English | MEDLINE | ID: mdl-32724153

ABSTRACT

TP53 mutations are implicated in the progression of mucinous borderline tumors (MBOT) to mucinous ovarian carcinomas (MOC). Optimized immunohistochemistry (IHC) for TP53 has been established as a proxy for the TP53 mutation status in other ovarian tumor types. We aimed to confirm the ability of TP53 IHC to predict TP53 mutation status in ovarian mucinous tumors and to evaluate the association of TP53 mutation status with survival among patients with MBOT and MOC. Tumor tissue from an initial cohort of 113 women with MBOT/MOC was stained with optimized IHC for TP53 using tissue microarrays (75.2%) or full sections (24.8%) and interpreted using established criteria as normal or abnormal (overexpression, complete absence, or cytoplasmic). Cases were considered concordant if abnormal IHC staining predicted deleterious TP53 mutations. Discordant tissue microarray cases were re-evaluated on full sections and interpretational criteria were refined. The initial cohort was expanded to a total of 165 MBOT and 424 MOC for the examination of the association of survival with TP53 mutation status, assessed either by TP53 IHC and/or sequencing. Initially, 82/113 (72.6%) cases were concordant using the established criteria. Refined criteria for overexpression to account for intratumoral heterogeneity and terminal differentiation improved concordance to 93.8% (106/113). In the expanded cohort, 19.4% (32/165) of MBOT showed evidence for TP53 mutation and this was associated with a higher risk of recurrence, disease-specific death, and all-cause mortality (overall survival: HR = 4.6, 95% CI 1.5-14.3, p = 0.0087). Within MOC, 61.1% (259/424) harbored a TP53 mutation, but this was not associated with survival (overall survival, p = 0.77). TP53 IHC is an accurate proxy for TP53 mutation status with refined interpretation criteria accounting for intratumoral heterogeneity and terminal differentiation in ovarian mucinous tumors. TP53 mutation status is an important biomarker to identify MBOT with a higher risk of mortality.


Subject(s)
Biomarkers, Tumor/genetics , DNA Mutational Analysis , Immunohistochemistry , Mutation , Neoplasms, Cystic, Mucinous, and Serous/genetics , Ovarian Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adult , Australia , Female , Humans , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/mortality , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , North America , Observer Variation , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Predictive Value of Tests , Prognosis , Reproducibility of Results , Risk Assessment , Risk Factors , Tissue Array Analysis , United Kingdom
7.
Cancers (Basel) ; 12(8)2020 Aug 08.
Article in English | MEDLINE | ID: mdl-32784519

ABSTRACT

Cancer cell lines are amongst the most important pre-clinical models. In the context of epithelial ovarian cancer, a highly heterogeneous disease with diverse subtypes, it is paramount to study a wide panel of models in order to draw a representative picture of the disease. As this lethal gynaecological malignancy has seen little improvement in overall survival in the last decade, it is all the more pressing to support future research with robust and diverse study models. Here, we describe ten novel spontaneously immortalized patient-derived ovarian cancer cell lines, detailing their respective mutational profiles and gene/biomarker expression patterns, as well as their in vitro and in vivo growth characteristics. Eight of the cell lines were classified as high-grade serous, while two were determined to be of the rarer mucinous and clear cell subtypes, respectively. Each of the ten cell lines presents a panel of characteristics reflective of diverse clinically relevant phenomena, including chemotherapeutic resistance, metastatic potential, and subtype-associated mutations and gene/protein expression profiles. Importantly, four cell lines formed subcutaneous tumors in mice, a key characteristic for pre-clinical drug testing. Our work thus contributes significantly to the available models for the study of ovarian cancer, supplying additional tools to better understand this complex disease.

8.
Int J Gynecol Pathol ; 39(2): 128-135, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30789501

ABSTRACT

The origin of serous endometrial intraepithelial carcinoma (SEIC) is debated, due to its premalignant and independently malignant nature. It often arises next to endometrial serous carcinoma (ESC), with a propensity for polypoid growth. We aimed to better characterize this discrepancy by analyzing the clinical, histologic, and immunohistochemical features of polypoid carcinoma associated with SEIC (P-SEIC), and compared them with usual endometrial serous carcinoma without SEIC (UESC). Consecutive patients with P-SEIC were recruited and compared with UESC controls from our institutional research center. Clinical, histologic, and immunohistochemical (IHC, ER, PR, P53, Napsin-A, WT1, P16) were analyzed. BRCA testing results and familial history were also extracted from clinical databases. Welch T test, Pearson χ, and Fisher exact test were performed in SPSS version 23. A total of 37 P-SEIC and 25 UESC were the basis of a case-control study. P-SEIC was associated with more bilateral ovarian involvement (P=0.026), yet showed lower rates of myometrial invasion (P=0.002). P-SEIC showed a statistically different IHC profile: p53+, p16+, ER+, PR+, and WT-1+, and high rates of Napsin-A, while UESC was p53+, p16+, WT-1-, Napsin-A-, with lower rates of ER and PR. We also identified 2 patients who received prophylactic salpingo-oophorectomy for BRCA mutations and who subsequently developed P-SEIC with its unique IHC pattern. Our results suggest different underlying expression profiles and possibly diverging molecular signatures between both P-SEIC and UESC. If confirmed in further molecular studies, it could lead to a distinct molecular subclass.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma in Situ/pathology , Cystadenocarcinoma, Serous/pathology , Endometrial Neoplasms/pathology , Aged , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Case-Control Studies , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Polyps
9.
Int J Cancer ; 146(7): 1800-1809, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31199510

ABSTRACT

Results of epidemiologic studies of physical activity and ovarian cancer risk are inconsistent. Few have attempted to measure physical activity over the lifetime or in specific age windows, which may better capture etiologically relevant exposures. We examined participation in moderate-to-vigorous recreational physical activity (MVPA) in relation to ovarian cancer risk. In a population-based case-control study conducted in Montreal, Canada from 2011 to 2016 (485 cases and 887 controls), information was collected on lifetime participation in various recreational physical activities, which was used to estimate MVPA for each participant. MVPA was represented as average energy expenditure over the lifetime and in specific age-periods in units of metabolic equivalents (METs)-hours per week. Odds ratios (OR) and 95% confidence intervals (CI) for the relation between average MVPA and ovarian cancer risk were estimated using multivariable logistic regression models. Confounding was assessed using directed acyclic graphs combined with a change-in-estimate approach. The adjusted OR (95% CI) for each 28.5 MET-hr/week increment of lifetime recreational MVPA was 1.11 (0.99-1.24) for ovarian cancer overall. ORs for individual age-periods were weaker. When examined by menopausal status, the OR (95% CI) for lifetime MVPA was 1.21 (1.00-1.45) for those diagnosed before menopause and 1.04 (0.89-1.21) for those diagnosed postmenopausally. The suggestive positive associations were stronger for invasive ovarian cancers and more specifically for high-grade serous carcinomas. These results do not support a reduced ovarian cancer risk associated with MVPA.


Subject(s)
Exercise , Leisure Activities , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/etiology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Humans , Middle Aged , Odds Ratio , Risk Assessment , Risk Factors , Young Adult
10.
Clin Cancer Res ; 26(5): 1009-1016, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31831561

ABSTRACT

PURPOSE: Platinum resistance in ovarian cancer is associated with epigenetic modifications. Hypomethylating agents (HMA) have been studied as carboplatin resensitizing agents in ovarian cancer. This randomized phase II trial compared guadecitabine, a second-generation HMA, and carboplatin (G+C) against second-line chemotherapy in women with measurable or detectable platinum-resistant ovarian cancer. PATIENTS AND METHODS: Patients received either G+C (guadecitabine 30 mg/m2 s.c. once-daily for 5 days and carboplatin) or treatment of choice (TC; topotecan, pegylated liposomal doxorubicin, paclitaxel, or gemcitabine) in 28-day cycles until progression or unacceptable toxicity. The primary endpoint was progression-free survival (PFS); secondary endpoints were RECIST v1.1 and CA-125 response rate, 6-month PFS, and overall survival (OS). RESULTS: Of 100 patients treated, 51 received G+C and 49 received TC, of which 27 crossed over to G+C. The study did not meet its primary endpoint as the median PFS was not statistically different between arms (16.3 weeks vs. 9.1 weeks in the G+C and TC groups, respectively; P = 0.07). However, the 6-month PFS rate was significantly higher in the G+C group (37% vs. 11% in TC group; P = 0.003). The incidence of grade 3 or higher toxicity was similar in G+C and TC groups (51% and 49%, respectively), with neutropenia and leukopenia being more frequent in the G+C group. CONCLUSIONS: Although this trial did not show superiority for PFS of G+C versus TC, the 6-month PFS increased in G+C treated patients. Further refinement of this strategy should focus on identification of predictive markers for patient selection.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Epigenesis, Genetic/drug effects , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Carboplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Female , Humans , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Patient Safety , Polyethylene Glycols/administration & dosage , Survival Rate , Topotecan/administration & dosage , Treatment Outcome , Gemcitabine
11.
Nat Commun ; 10(1): 3935, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477716

ABSTRACT

Mucinous ovarian carcinoma (MOC) is a unique subtype of ovarian cancer with an uncertain etiology, including whether it genuinely arises at the ovary or is metastatic disease from other organs. In addition, the molecular drivers of invasive progression, high-grade and metastatic disease are poorly defined. We perform genetic analysis of MOC across all histological grades, including benign and borderline mucinous ovarian tumors, and compare these to tumors from other potential extra-ovarian sites of origin. Here we show that MOC is distinct from tumors from other sites and supports a progressive model of evolution from borderline precursors to high-grade invasive MOC. Key drivers of progression identified are TP53 mutation and copy number aberrations, including a notable amplicon on 9p13. High copy number aberration burden is associated with worse prognosis in MOC. Our data conclusively demonstrate that MOC arise from benign and borderline precursors at the ovary and are not extra-ovarian metastases.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Carcinoma, Ovarian Epithelial/genetics , Gene Expression Profiling/methods , Ovarian Neoplasms/genetics , Adenocarcinoma, Mucinous/classification , Adenocarcinoma, Mucinous/metabolism , Carcinoma, Ovarian Epithelial/classification , Carcinoma, Ovarian Epithelial/metabolism , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Ovarian Neoplasms/classification , Ovarian Neoplasms/metabolism , Sequence Analysis, DNA/methods , Survival Analysis
12.
J Pathol Clin Res ; 5(3): 177-188, 2019 07.
Article in English | MEDLINE | ID: mdl-30924313

ABSTRACT

The aim of this study was to describe the expression of special AT-rich sequence-binding protein 2 (SATB2) in ovarian endometrioid carcinoma (EC). SATB2 is a nuclear matrix-associated transcription factor that is associated with abnormal expression in certain cancers but has not been reported for ovarian carcinoma. SATB2 mRNA and protein expression was first assessed in a pilot cohort of 26 samples by Affymetrix microarray and by routine immunohistochemistry on a small tissue microarray. A large multicenter validation cohort representing the well-characterized cases of 235 ovarian EC from the Canadian Ovarian Experimental Unified Resource (COEUR) was then used to validate this result and to assess the prognostic impact of SATB2 expression. SATB2 staining was scored as negative, weak, moderate, and strong intensity, and by percentage of stained cells. No SATB2 expression was observed in clear cell carcinomas but 10% (n = 3) of the ECs in the pilot cohort showed SATB2 expression. In the validation cohort, strong expression was observed in 11% of ECs, while weak or moderate expression levels were detected in 12% of cases. Evaluation of SATB2 expression with clinicopathological parameters revealed an association with patient age and Federation International of Gynecology and Obstetrics grade but not with disease stage or postoperative residual disease. Any expression of SATB2, independent of intensity, was also associated with longer survival and improved progression-free survival with hazard ratio (HR) = 0.14 (95% CI 0.03-0.56) and HR = 0.16 (95% CI 0.02-1.24) respectively. A greater beneficial effect was observed in patients with stage III/IV disease compared to patients with stage I/II disease. Furthermore, direct comparison of SATB2 with other reported prognostic biomarkers such as progesterone receptor, CDX2 and ß-catenin within this cohort showed that SATB2 had the strongest association with survival. Given the current lack of accurate prognostic factors for these patients, SATB2 has promising clinical utility and warrants further study.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Endometrioid/pathology , Carcinoma, Ovarian Epithelial/pathology , Matrix Attachment Region Binding Proteins/biosynthesis , Transcription Factors/biosynthesis , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/mortality , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/mortality , Cohort Studies , Female , Humans , Matrix Attachment Region Binding Proteins/analysis , Middle Aged , Pilot Projects , Prognosis , Progression-Free Survival , Transcription Factors/analysis
13.
Cancer Epidemiol Biomarkers Prev ; 28(5): 987-995, 2019 05.
Article in English | MEDLINE | ID: mdl-30842128

ABSTRACT

BACKGROUND: Shift work causing circadian disruption is classified as a "probable carcinogen" and may contribute to the pathogenesis of hormone-sensitive cancers. This study investigated shift work exposure in relation to epithelial ovarian cancer (EOC) risk. METHODS: In a population-based case-control study with 496 EOC cases and 906 controls, lifetime occupational histories were collected and used to calculate cumulative years of shift work exposure, average number of night shifts per month, and average number of consecutive night shifts per month. ORs and 95% confidence intervals (CI) for associations with EOC risk were estimated using logistic regression. Associations were also examined according to chronotype and menopausal status. RESULTS: More than half of the cases (53.4%) and controls (51.7%) worked evening and/or night shifts. There was no clear pattern of increasing EOC risk with increasing years of shift work; the adjusted OR of EOC comparing the highest shift work category versus never working shift work was 1.20 (95% CI, 0.89-1.63). This association was more pronounced among those self-identified as having a "morning" chronotype (OR, 1.64; 95% CI, 1.01-2.65). Associations did not greatly differ by menopausal status. CONCLUSIONS: These results do not strongly demonstrate a relationship between shift work and EOC risk. IMPACT: This study collected detailed shift work information and examined shift work patterns according to shift times and schedules. The findings highlight that chronotype should be considered in studies of shift work as an exposure.


Subject(s)
Carcinoma, Ovarian Epithelial/epidemiology , Ovarian Neoplasms/epidemiology , Shift Work Schedule/statistics & numerical data , Adult , Aged , Canada/epidemiology , Carcinoma, Ovarian Epithelial/etiology , Case-Control Studies , Circadian Rhythm , Female , Follow-Up Studies , Humans , Incidence , Middle Aged , Ovarian Neoplasms/etiology , Prognosis , Risk Factors , Shift Work Schedule/adverse effects , Surveys and Questionnaires , Work Schedule Tolerance , Young Adult
14.
BMC Cancer ; 18(1): 347, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29587661

ABSTRACT

BACKGROUND: Ovarian carcinoma is the most lethal gynecological malignancy due to early dissemination and acquired resistance to platinum-based chemotherapy. Reliable markers that are independent and complementary to clinical parameters are needed to improve the management of patients with this disease. The Canadian Ovarian Experimental Unified Resource (COEUR) provides researchers with biological material and associated clinical data to conduct biomarker validation studies. Using standards defined by the Canadian Tissue Repository Network (CTRNet), we have previously demonstrated the quality of the biological material from this resource. Here we describe the clinical characteristics of the COEUR cohort. METHODS: With support from 12 Canadian ovarian cancer biobanks in Canada, we created a central retrospective cohort comprised of more than 2000 patient tissue samples with associated clinical data, including 1246 high-grade serous, 102 low-grade serous, 295 endometrioid, 259 clear cell and 89 mucinous carcinoma histotypes. A two-step reclassification process was applied to assure contemporary histological classification (histotyping). For each histotypes individually, we evaluated the association between the known clinico-pathological parameters (stage, cytoreduction, chemotherapy treatment, BRCA1 and BRCA2 mutation) and patient outcome by using Kaplan-Meier and Cox proportional hazard regression analyses. RESULTS: The median follow-up time of the cohort was 45 months and the 5-year survival rate for patients with high-grade serous carcinomas was 34%, in contrast to endometrioid carcinomas with 80% at 5 years. Survival profiles differed by histotype when stratified by stage or cytoreduction. Women with mucinous or clear cell carcinomas at advanced stage or with non-optimally debulked disease had the worst outcomes. In high-grade serous carcinoma, we observed significant association with longer survival in women harboring BRCA1 or BRCA2 mutation as compared to patients without detectable mutation. CONCLUSIONS: Our results show the expected survival rates, as compared with current literature, in each histotype suggesting that the cohort is an unbiased representation of the five major histotypes. COEUR, a one stop comprehensive biorepository, has collected mature outcome data and relevant clinical data in a comprehensive manner allowing stratified analysis.


Subject(s)
Biomarkers, Tumor , Ovarian Neoplasms/diagnosis , Aged , Biological Specimen Banks , Canada , Cohort Studies , Female , Genes, BRCA1 , Genes, BRCA2 , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Prognosis , Proportional Hazards Models
15.
J Biomed Opt ; 22(7): 76012, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28727868

ABSTRACT

The majority of high-grade serous ovarian cancers is now believed to originate in the fallopian tubes. Therefore, current practices include the pathological examination of excised fallopian tubes. Detection of tumors in the fallopian tubes using current clinical approaches remains difficult but is of critical importance to achieve accurate staging and diagnosis. Here, we present an intraoperative imaging system for the detection of human fallopian tube lesions. The system is based on optical coherence tomography (OCT) to access subepithelial tissue architecture. To demonstrate that OCT could identify lesions, we analyzed 180 OCT volumes taken from five different ovarian lesions and from healthy fallopian tubes, and compared them to standard pathological review. We demonstrated that qualitative features could be matched to pathological conditions. We then determined the feasibility of intraluminal imaging of intact human fallopian tubes by building a dedicated endoscopic single-fiber OCT probe to access the mucosal layer inside freshly excised specimens from five patients undergoing prophylactic surgeries. The probe insertion into the lumen acquired images over the entire length of the tubes without damaging the mucosa, providing the first OCT images of intact human fallopian tubes.


Subject(s)
Fallopian Tubes/diagnostic imaging , Imaging, Three-Dimensional/instrumentation , Ovarian Neoplasms/diagnostic imaging , Tomography, Optical Coherence , Female , Humans , Mucous Membrane/pathology
16.
Cancer Causes Control ; 28(5): 393-403, 2017 May.
Article in English | MEDLINE | ID: mdl-28102526

ABSTRACT

PURPOSE: Hormone-related factors have been associated with ovarian cancer, the strongest being parity and oral contraceptive use. Given reductions in birth rates and increases in oral contraceptive use over time, associations in more recent birth cohorts may differ. Furthermore, consideration of ovarian cancer heterogeneity (i.e., Type I/II invasive cancers) may contribute to a better understanding of etiology. We examined hormone-related factors in relation to ovarian cancer risk overall, for Type I and Type II cancers, as well as borderline tumors. METHODS: A population-based case-control study was carried out in Montreal, Canada from 2011 to 2016, including 496 cases and 908 controls. For each hormone-related variable, adjusted odds ratios (OR) and 95% confidence intervals (CI) were estimated using logistic regression for ovarian cancer overall, and using polytomous logistic regression for associations by tumor behavior and ovarian cancer type. RESULTS: Parity was inversely associated with risk overall and by tumor behavior and type, with a stronger OR (95% CI) for Type I [0.09 (0.04-0.24) for ≥3 full-term births vs. nulliparity] vs. Type II [0.66 (0.43-1.02)] invasive cancers; the OR (95% CI) for borderline tumors was 0.41 (0.22-0.77). Oral contraceptive ever use was not associated with risk overall, but ≥10 years of use vs. never use reduced risk, particularly for invasive cancers. A history of endometriosis was most strongly associated with Type I cancers. Associations with other factors were less clear. CONCLUSIONS: These results suggest that associations with some hormone-related factors may differ between borderline and invasive Type I and II ovarian cancers.


Subject(s)
Contraceptives, Oral/adverse effects , Ovarian Neoplasms/etiology , Parity , Reproductive History , Adolescent , Adult , Aged , Canada , Case-Control Studies , Child , Female , Humans , Middle Aged , Ovarian Neoplasms/epidemiology , Pregnancy , Prevalence , Risk Factors , Young Adult
17.
Hum Pathol ; 62: 33-39, 2017 04.
Article in English | MEDLINE | ID: mdl-27864117

ABSTRACT

Although many studies have evaluated the impact of mismatch repair protein loss of expression (MMR LOE) or microcystic, elongated, and fragmented (MELF) pattern of myometrial invasion as individual factors in endometrial cancer, we analyzed the combined impact of both. We reviewed every case of International Federation of Gynecology and Obstetrics (FIGO) grade 1 endometrioid endometrial cancers (EECs) from our institution, between 2011 and 2015, that had a sentinel lymph node biopsy and/or a lymphadenectomy, and examined the following data: age, myometrial infiltration, MELF infiltration, lymphovascular space invasion, and lymph node status. These cases were then grouped according to the absence of lymph node metastases, the presence of isolated tumor cell (ITC) lymph node metastases, or the presence of non-ITC metastases. Among the 127 cases that were in our study, 105 patients did not have nodal metastases, whereas 22 patients showed metastases, of which 11 were ITC. MMR LOE was only significantly associated with a higher odds ratio (OR) of metastases (OR, 7.44; P < .001). MELF was only associated with a higher OR of ITC-pattern metastases (OR, 32.3; P < .001). This study distinguished the effects of MELF and MMR LOE on the risk of metastases in FIGO grade 1 EEC. Further research on the clinical impact of MELF and ITC-pattern metastases is warranted to better guide clinicians on the management of patients with FIGO grade 1 EEC harboring such characteristics, which are still considered low-risk cancer.


Subject(s)
Carcinoma, Endometrioid/secondary , Cell Movement , Endometrial Neoplasms/pathology , Myometrium/pathology , Base Pair Mismatch , Carcinoma, Endometrioid/chemistry , Carcinoma, Endometrioid/surgery , DNA Repair Enzymes/analysis , Endometrial Neoplasms/chemistry , Endometrial Neoplasms/surgery , Female , Humans , Hysterectomy , Immunohistochemistry , Logistic Models , Lymph Node Excision , Lymphatic Metastasis , Middle Aged , Multivariate Analysis , Neoplasm Grading , Neoplasm Invasiveness , Odds Ratio , Predictive Value of Tests , Quebec , Retrospective Studies , Risk Factors , Sentinel Lymph Node Biopsy
18.
Gynecol Oncol ; 143(1): 27-34, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27546885

ABSTRACT

PURPOSE: Trebananib, a peptibody that blocks binding of angiopoietin-1 and -2 to Tie2, significantly prolonged progression-free survival (PFS) in patients with recurrent epithelial ovarian cancer in the phase 3 TRINOVA-1 study. We report overall survival (OS) in the intent-to-treat population and clinically relevant subgroups and time to second disease progression (PFS-2). PATIENTS AND METHODS: Women with recurrent disease (platinum-free interval<12months) were randomized to receive intravenous paclitaxel 80mg/m(2) (3weeks on/1week off) plus intravenous trebananib 15mg/kg or placebo, weekly. OS in the intent-to-treat population was a key secondary endpoint. Exploratory analysis of PFS-2 was conducted according to guidance by the European Medicines Agency. RESULTS: Median OS was not significantly improved with trebananib compared with placebo (19.3 versus 18.3months; HR, 0.95; 95% CI, 0.81-1.11; P=0.52) in the intent-to-treat population (n=919). In subgroup analysis, trebananib improved median OS compared with placebo (14.5 versus 12.3months; HR, 0.72; 95% CI, 0.55-0.93; P=0.011) in patients with ascites at baseline (n=295). In the intent-to-treat population, trebananib significantly improved median PFS-2 compared with placebo (12.5 versus 10.9months; HR, 0.85; 95% CI, 0.74-0.98; P=0.024). The incidence and type of adverse events in this updated analysis was consistent with that described in the primary analysis; no new safety signals were detected. CONCLUSIONS: OS was not significantly longer in the intent-to-treat population, although there was an improvement in OS in patients with ascites receiving trebananib. PFS-2 confirmed that the PFS benefit associated with trebananib was maintained through the second disease progression independent of the choice of subsequent therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ascites/etiology , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Double-Blind Method , Female , Humans , Middle Aged , Neoplasm Recurrence, Local/mortality , Ovarian Neoplasms/complications , Ovarian Neoplasms/mortality , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects
19.
Oncol Lett ; 9(6): 2787-2790, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26137147

ABSTRACT

French Canadian families with breast cancer and breast-ovarian cancer syndrome harbor specific BRCA1, BRCA2 and PALB2 germline mutations, which have been attributed to common founders. Mutations in these genes confer an increased risk to breast and ovarian cancers, and have been identified to play a role in and directly interact with the common homologous recombination DNA repair pathways. Our previous study described the case of a female diagnosed with breast cancer at 45 years old, who harbored the PALB2:c.2323C>T [p.Q775X] and BRCA2:c.9004G>A [p.E3002K] germline mutations, which have been found to recur in the French Canadian cancer families. As the frequency of double heterozygous carriers of breast-ovarian cancer susceptibility alleles is unknown, and due to the possibility that there may be implications for genetic counseling and management for these carriers, the present study investigated the co-occurrence of BRCA1/BRCA2 and PALB2 mutations in the French Canadian cancer families. The PALB2:c.2323C>T [p.Q775X] mutation, which is the only PALB2 mutation to have been identified in French Canadian cancer families, was screened in 214 breast cancer cases and 22 breast-ovarian cancer cases from 114 BRCA1/BRCA2 mutation-positive French Canadian breast cancer (n=61) and breast-ovarian cancer (n=53) families using a tailored polymerase chain reaction-based TaqMan® SNP Genotyping Assay. No additional PALB2:c.2323C>T [p.Q775X] mutation carriers were identified among the BRCA1/BRCA2 mutation carriers. The results suggest that carriers of the PALB2:c.2323C>T [p.Q775X] mutation rarely co-occur in French Canadian breast cancer and breast-ovarian cancer families harboring BRCA1 or BRCA2 mutations.

20.
Int J Gynecol Cancer ; 24(9 Suppl 3): S102-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25341572

ABSTRACT

Small cell carcinoma of the cervix (SCCC) is a rare histological entity of uterine cervical cancer. Compared with other common histological types, squamous cell carcinoma or adenocarcinoma, the outcome of SCCC is poor because of the high incidence of nodal or distant metastasis even with early stage. In this review, current consensus of epidemiology, pathology, and initial treatment for SCCC will be discussed.


Subject(s)
Carcinoma, Small Cell/pathology , Medical Oncology , Practice Guidelines as Topic , Uterine Cervical Neoplasms/pathology , Carcinoma, Small Cell/therapy , Combined Modality Therapy , Consensus , Female , Humans , Societies, Medical , Uterine Cervical Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...