Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cell Sci ; 136(19)2023 10 01.
Article in English | MEDLINE | ID: mdl-37701987

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) exhibits elevated levels of autophagy, which promote tumor progression and treatment resistance. ATG4B is an autophagy-related cysteine protease under consideration as a potential therapeutic target, but it is largely unexplored in PDAC. Here, we investigated the clinical and functional relevance of ATG4B expression in PDAC. Using two PDAC patient cohorts, we found that low ATG4B mRNA or protein expression is associated with worse patient survival outcomes, poorly differentiated PDAC tumors and a lack of survival benefit from adjuvant chemotherapy. In PDAC cell lines, ATG4B knockout reduced proliferation, abolished processing of LC3B (also known as MAP1LC3B), and reduced GABARAP and GABARAPL1 levels, but increased ATG4A levels. ATG4B and ATG4A double knockout lines displayed a further reduction in proliferation, characterized by delays in G1-S phase transition and mitosis. Pro-LC3B accumulated aberrantly at the centrosome with a concomitant increase in centrosomal proteins PCM1 and CEP131, which was rescued by exogenous ATG4B. The two-stage cell cycle defects following ATG4B and ATG4A loss have important therapeutic implications for PDAC.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Pancreatic Neoplasms/genetics , Autophagy/genetics , Cell Line, Tumor , Cell Cycle/genetics , Cell Proliferation/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms
2.
J Pathol ; 254(3): 254-264, 2021 07.
Article in English | MEDLINE | ID: mdl-33797756

ABSTRACT

Hereditary diffuse gastric cancer (HDGC) is a cancer syndrome caused by germline variants in CDH1, the gene encoding the cell-cell adhesion molecule E-cadherin. Loss of E-cadherin in cancer is associated with cellular dedifferentiation and poor prognosis, but the mechanisms through which CDH1 loss initiates HDGC are not known. Using single-cell RNA sequencing, we explored the transcriptional landscape of a murine organoid model of HDGC to characterize the impact of CDH1 loss in early tumourigenesis. Progenitor populations of stratified squamous and simple columnar epithelium, characteristic of the mouse stomach, showed lineage-specific transcriptional programs. Cdh1 inactivation resulted in shifts along the squamous differentiation trajectory associated with aberrant expression of genes central to gastrointestinal epithelial differentiation. Cytokeratin 7 (CK7), encoded by the differentiation-dependent gene Krt7, was a specific marker for early neoplastic lesions in CDH1 carriers. Our findings suggest that deregulation of developmental transcriptional programs may precede malignancy in HDGC. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cadherins/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Predisposition to Disease/genetics , Stomach Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Mice , Mice, Transgenic , Organoids , Single-Cell Analysis , Stomach Neoplasms/pathology , Transcriptome
3.
Int J Cancer ; 148(2): 481-491, 2021 01 15.
Article in English | MEDLINE | ID: mdl-32955725

ABSTRACT

The mixture of epithelial and stromal components in pancreatic ductal adenocarcinoma (PDAC) may confound sequencing-based studies of tumor gene expression. Virtual microdissection has been suggested as a bioinformatics approach to segment the aforementioned components, and subsequent prognostic gene sets have emerged from this research. We examined the prognostic signature from the epithelial gene set of one such study using laser capture microdissected (LCM) epithelial samples. We also examined this gene set in matched stromal samples to determine whether prognostic findings were specific to the epithelium. LCM samples from 48 long-term and 48 short-term PDAC survivors were obtained. The resultant epithelial and stromal components were subjected to direct mRNA quantification using a 49 gene published PDAC classifier. Component-specific unsupervised hierarchical clustering was used to derive groups and survival differences were quantified. Immunohistochemical validation of particular genes was performed in an independent cohort. Clustering in the epithelial component yielded prognostic differences in univariable analysis (P = .02), but those differences were not significant when controlled for other clinicopathologic covariates (P = .06). Clustering in the stromal component yielded prognostic differences that persisted in the presence of other clinicopathologic covariates (P = .0005). Validation of selected genes in the epithelium (KRT6A-negative prognostic [P = .004]) and stroma (LY6D-improved prognostic [P = .01] and CTSV-negative prognostic [P = .0002]) demonstrated statistical independence in multivariable analysis. Although the genes used in this study were originally identified as being representative of the epithelial component of PDAC, their expression in the stroma appears to provide additional information that may aid in improved prognostication.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cluster Analysis , Cohort Studies , Epithelial Cells/pathology , Formaldehyde , Gene Expression , Humans , Laser Capture Microdissection , Lymph Nodes/pathology , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Paraffin Embedding , Peripheral Nerves/pathology , Prognosis , Stromal Cells/pathology , Survival Analysis , Tissue Fixation
4.
Gastroenterology ; 157(3): 823-837, 2019 09.
Article in English | MEDLINE | ID: mdl-31078621

ABSTRACT

BACKGROUND & AIMS: Most pancreatic ductal adenocarcinomas (PDACs) express an activated form of KRAS, become hypoxic and dysplastic, and are refractory to chemo and radiation therapies. To survive in the hypoxic environment, PDAC cells upregulate enzymes and transporters involved in pH regulation, including the extracellular facing carbonic anhydrase 9 (CA9). We evaluated the effect of blocking CA9, in combination with administration of gemcitabine, in mouse models of pancreatic cancer. METHODS: We knocked down expression of KRAS in human (PK-8 and PK-1) PDAC cells with small hairpin RNAs. Human and mouse (KrasG12D/Pdx1-Cre/Tp53/RosaYFP) PDAC cells were incubated with inhibitors of MEK (trametinib) or extracellular signal-regulated kinase (ERK), and some cells were cultured under hypoxic conditions. We measured levels and stability of the hypoxia-inducible factor 1 subunit alpha (HIF1A), endothelial PAS domain 1 protein (EPAS1, also called HIF2A), CA9, solute carrier family 16 member 4 (SLC16A4, also called MCT4), and SLC2A1 (also called GLUT1) by immunoblot analyses. We analyzed intracellular pH (pHi) and extracellular metabolic flux. We knocked down expression of CA9 in PDAC cells, or inhibited CA9 with SLC-0111, incubated them with gemcitabine, and assessed pHi, metabolic flux, and cytotoxicity under normoxic and hypoxic conditions. Cells were also injected into either immune-compromised or immune-competent mice and growth of xenograft tumors was assessed. Tumor fragments derived from patients with PDAC were surgically ligated to the pancreas of mice and the growth of tumors was assessed. We performed tissue microarray analyses of 205 human PDAC samples to measure levels of CA9 and associated expression of genes that regulate hypoxia with outcomes of patients using the Cancer Genome Atlas database. RESULTS: Under hypoxic conditions, PDAC cells had increased levels of HIF1A and HIF2A, upregulated expression of CA9, and activated glycolysis. Knockdown of KRAS in PDAC cells, or incubation with trametinib, reduced the posttranscriptional stabilization of HIF1A and HIF2A, upregulation of CA9, pHi, and glycolysis in response to hypoxia. CA9 was expressed by 66% of PDAC samples analyzed; high expression of genes associated with metabolic adaptation to hypoxia, including CA9, correlated with significantly reduced survival times of patients. Knockdown or pharmacologic inhibition of CA9 in PDAC cells significantly reduced pHi in cells under hypoxic conditions, decreased gemcitabine-induced glycolysis, and increased their sensitivity to gemcitabine. PDAC cells with knockdown of CA9 formed smaller xenograft tumors in mice, and injection of gemcitabine inhibited tumor growth and significantly increased survival times of mice. In mice with xenograft tumors grown from human PDAC cells, oral administration of SLC-0111 and injection of gemcitabine increased intratumor acidosis and increased cell death. These tumors, and tumors grown from PDAC patient-derived tumor fragments, grew more slowly than xenograft tumors in mice given control agents, resulting in longer survival times. In KrasG12D/Pdx1-Cre/Tp53/RosaYFP genetically modified mice, oral administration of SLC-0111 and injection of gemcitabine reduced numbers of B cells in tumors. CONCLUSIONS: In response to hypoxia, PDAC cells that express activated KRAS increase expression of CA9, via stabilization of HIF1A and HIF2A, to regulate pH and glycolysis. Disruption of this pathway slows growth of PDAC xenograft tumors in mice and might be developed for treatment of pancreatic cancer.


Subject(s)
Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Microenvironment , Animals , Antigens, Neoplasm/genetics , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase Inhibitors/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Glycolysis/drug effects , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Phenylurea Compounds/pharmacology , Signal Transduction , Sulfonamides/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
5.
Histopathology ; 77(1): 46-54, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32413172

ABSTRACT

AIMS: Undifferentiated carcinoma refers to an epithelial malignancy that lacks morphological evidence of differentiation. Recent studies have implicated the loss of constitutively expressed switch/sucrose non-fermenting (SWI/SNF) complex subunits in undifferentiated carcinomas of the gastrointestinal tract and other sites. In this study we examine the expression of SWI/SNF and mismatch repair (MMR) proteins in a series of undifferentiated carcinomas from the gastrointestinal tract and the pancreas. METHODS AND RESULTS: We searched pathology databases from four Canadian health centres for primary undifferentiated carcinoma from gastrointestinal and pancreatic resection specimens. Upon review of 31 cases, 19 were confirmed to be undifferentiated carcinomas (eight colonic, six gastric, three pancreatic, one appendiceal and one duodenal). Immunohistochemical analysis of SMARCA4, SMARCA2, SMARCB1, ARID1A, ARID1B, MSH2, MSH6, MLH1 and PMS2 was performed on whole sections. Five of 19 (26%) showed loss of core SWI/SNF proteins (two loss of SMARCA4, one loss of SMARCB1 and two concurrent loss of ARID1A and ARID1B). SMARCA4, SMARCB1, or ARID1A/ARID1B-deficient undifferentiated carcinoma consistently exhibited sheet-like growth pattern, with cellular discohesion and rhabdoid morphology. Nine of 17 undifferentiated carcinomas tested were MMR-deficient by immunohistochemistry. In comparison, none of the 12 poorly differentiated carcinomas that were originally diagnosed as undifferentiated carcinomas showed loss of SMARCA4, SMARCA2, SMARCB1 or ARID1B. CONCLUSIONS: Undifferentiated gastrointestinal/pancreatic carcinomas show frequent loss of expression of SWI/SNF complex proteins. The loss of these core components of SWI/SNF complex may contribute to the arrest of cellular differentiation, resulting in the undifferentiated histology and aggressive clinical behaviour.


Subject(s)
Biomarkers, Tumor/analysis , Gastrointestinal Neoplasms/pathology , Pancreatic Neoplasms/pathology , Transcription Factors/biosynthesis , Adult , Aged , Aged, 80 and over , Female , Gastrointestinal Neoplasms/metabolism , Humans , Male , Middle Aged , Pancreatic Neoplasms/metabolism , Transcription Factors/analysis , Pancreatic Neoplasms
6.
Health Expect ; 23(4): 884-892, 2020 08.
Article in English | MEDLINE | ID: mdl-32338425

ABSTRACT

BACKGROUND: Multi-gene panel testing is replacing single-gene testing for patients with suspected hereditary cancer syndromes. The detection of a hereditary cancer syndrome allows tested individuals to initiate enhanced primary and secondary prevention efforts-where available-with a view to reduce disease burden. Current policy prevents testing programmes from communicating genetic test results with potentially affected family members, yet it is well documented that tested individuals face multiple challenges in initiating such discussions with relatives. OBJECTIVE: In response to this challenge, we sought patient recommendations about how to improve genetic risk communication to enhance interfamilial discussions about primary and secondary disease prevention. DESIGN: We conducted 25 semi-structured interviews with individuals who received genetic testing through British Columbia's Hereditary Cancer Program between 2017 and 2018. Interviews were professionally transcribed and analysed using a constant comparative approach. RESULTS: Participants described difficulty engaging in conversations with relatives who were resistant to receiving genetic risk information, when communicating with younger relatives and where participants reported strained familial relationships. Participants recommended that testing facilities provide a summary of results and implications and that resources be made available to prepare patients for challenging discussions with family members. DISCUSSION: Our study demonstrates that individuals undergoing genetic testing for suspected hereditary cancer syndromes would benefit from additional supportive resources alongside genetic counselling. Providing this on-going support will enhance the accurate and transparent communication of risk to facilitate the uptake of cascade testing and enhanced prevention strategies.


Subject(s)
Genetic Testing , Neoplastic Syndromes, Hereditary , Communication , Family , Genetic Counseling , Genetic Predisposition to Disease , Humans , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/genetics
7.
BMC Cancer ; 17(1): 618, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28870260

ABSTRACT

BACKGROUND: Programmed cell death 1 (PD1) inhibitors have recently shown promising anti-cancer effects in a number of solid tumor types. A predictive biomarker to this class of drugs has not been clearly identified; however, overexpression of the PD1 ligand (PD-L1) has shown particular promise in lung adenocarcinoma. In this study, we explore the staining characteristics, prevalence, and clinico-molecular correlates of PD-L1 overexpression in pancreatic ductal adenocarcinoma (PDAC). METHODS: A tissue microarray (TMA) was constructed from cases of resected PDAC. PD-L1 immunohistochemistry (IHC) was performed using the SP142 primary antibody. Immunohistochemical assessment for deficient mismatch repair status (MMRd), CD3 and CD8 were performed. All biomarkers were assessed independently by two anatomical pathologists and consensus achieved on all cases. Survival analysis was performed using three thresholds (> = 1%, >5% and >10%) for tumor cell membrane staining. RESULTS: Two-hundred fifty-two cases were included in the TMA and evaluable by IHC. Thirty-one (12%), 17 (7%), 12(5%) cases were positive at percentage cut offs of >0, >5, and >10% respectively. Increased PD-L1 expression was associated with inferior prognosis (p = 0.0367). No statistically significant association was identified between PD-L1 status and MMR status or tumor infiltrating lymphocytes. CONCLUSIONS: This data suggests that there is an inverse relationship between PD-L1 expression and disease specific survival times in resected PDAC. Consequently, this association may represent a phenotype where increased PD-L1 expression has an effect on tumor biology and could therefore identify a subgroup where PD1 blockade could have enhanced effectiveness.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Aged , B7-H1 Antigen/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Chemotherapy, Adjuvant , DNA Mismatch Repair , Female , Gene Expression , Humans , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prognosis , Proportional Hazards Models , Tissue Array Analysis
8.
N Engl J Med ; 366(3): 234-42, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22187960

ABSTRACT

BACKGROUND: Germline truncating mutations in DICER1, an endoribonuclease in the RNase III family that is essential for processing microRNAs, have been observed in families with the pleuropulmonary blastoma-family tumor and dysplasia syndrome. Mutation carriers are at risk for nonepithelial ovarian tumors, notably sex cord-stromal tumors. METHODS: We sequenced the whole transcriptomes or exomes of 14 nonepithelial ovarian tumors and noted closely clustered mutations in the region of DICER1 encoding the RNase IIIb domain of DICER1 in four samples. We then sequenced this region of DICER1 in additional ovarian tumors and in certain other tumors and queried the effect of the mutations on the enzymatic activity of DICER1 using in vitro RNA cleavage assays. RESULTS: DICER1 mutations in the RNase IIIb domain were found in 30 of 102 nonepithelial ovarian tumors (29%), predominantly in Sertoli-Leydig cell tumors (26 of 43, or 60%), including 4 tumors with additional germline DICER1 mutations. These mutations were restricted to codons encoding metal-binding sites within the RNase IIIb catalytic centers, which are critical for microRNA interaction and cleavage, and were somatic in all 16 samples in which germline DNA was available for testing. We also detected mutations in 1 of 14 nonseminomatous testicular germ-cell tumors, in 2 of 5 embryonal rhabdomyosarcomas, and in 1 of 266 epithelial ovarian and endometrial carcinomas. The mutant DICER1 proteins had reduced RNase IIIb activity but retained RNase IIIa activity. CONCLUSIONS: Somatic missense mutations affecting the RNase IIIb domain of DICER1 are common in nonepithelial ovarian tumors. These mutations do not obliterate DICER1 function but alter it in specific cell types, a novel mechanism through which perturbation of microRNA processing may be oncogenic. (Funded by the Terry Fox Research Institute and others.).


Subject(s)
DEAD-box RNA Helicases/genetics , Mutation, Missense , Ovarian Neoplasms/genetics , Ribonuclease III/genetics , Sertoli-Leydig Cell Tumor/genetics , Carcinosarcoma/genetics , Female , Gene Expression , Gene Expression Profiling , Germ-Line Mutation , Humans , MicroRNAs/metabolism , Neoplasms, Germ Cell and Embryonal/genetics , Rhabdomyosarcoma/genetics , Sequence Analysis, DNA
9.
Mod Pathol ; 28(10): 1383-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26226846

ABSTRACT

Deficiencies in DNA mismatch repair have been associated with inferior response to 5-FU in colorectal cancer. Pancreatic ductal adenocarcinoma is similarly treated with pyrimidine analogs, yet the predictive value of mismatch repair status for response to these agents has not been examined in this malignancy. A tissue microarray with associated clinical outcome, comprising 254 resected pancreatic ductal adenocarcinoma patients was stained for four mismatch repair proteins (MLH1, MSH2, MSH6 and PMS2). Mismatch repair deficiency and proficiency was determined by the absence or presence of uniform nuclear staining in tumor cells, respectively. Cases identified as mismatch repair deficient on the tissue microarray were confirmed by immunohistochemistry on whole slide sections. Of the 265 cases, 78 (29%) received adjuvant treatment with a pyrimidine analog and 41 (15%) showed a mismatch repair-deficient immunoprofile. Multivariable disease-specific survival in the mismatch repair-proficient cohort demonstrated that adjuvant chemotherapy, regional lymph-node status, gender, and the presence of tumor budding were significant independent prognostic variables (P≤0.04); however, none of the eight clinico-pathologic covariates examined in the mismatch repair-deficient cohort were of independent prognostic significance. Univariable assessment of disease-specific survival revealed an almost identical survival profile for both treated and untreated patients with a mismatch repair-deficient profile, while treatment in the mismatch repair-proficient cohort conferred a greater than 10-month median disease-specific survival advantage over their untreated counterparts (P=0.0018). In this cohort, adjuvant chemotherapy with a pyrimidine analog conferred no survival advantage to mismatch repair-deficient pancreatic ductal adenocarcinoma patients. Mismatch repair immunoprofiling is a feasible predictive marker in pancreatic ductal adenocarcinoma patients, and further prospective evaluation of this finding is warranted.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/genetics , DNA Mismatch Repair/genetics , Drug Resistance, Neoplasm/genetics , Pancreatic Neoplasms/genetics , Aged , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Chemotherapy, Adjuvant , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Proportional Hazards Models , Tissue Array Analysis
10.
Mod Pathol ; 27(2): 255-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23887303

ABSTRACT

BAF250a (ARID1A) loss is a frequent event in high-grade endometrial cancers. It has been proposed that ARID1A is a driver gene, with ARID1A mutations occurring secondary to deregulated mismatch repair mechanism in gastric cancers, representing an alternative oncogenic pathway to p53 alteration. The prognostic significance of ARID1A loss is controversial. In this study, we investigated the frequency of BAF250a immunohistochemical loss in a cohort of high-grade endometrial cancers (n=190) and correlated it with mismatch repair (hMLH1, hMSH2, hMSH6, and hPMS2) and p53 protein expression. The 190 cases consisted of 82 high-grade endometrioid, 88 serous, 10 clear cell, and 10 mixed (carcinosarcomas and mixed histology). There was BAF250a loss in 55/190 (29%) cancers, most commonly in high-grade endometrioid carcinomas (46 vs 9% in serous carcinomas, P<0.0001). Loss of any mismatch repair proteins was observed in 63/190 (33%) cancers, most commonly in high-grade endometrioid carcinomas (57 vs 10% in serous carcinomas, P<0.0001). Aberrant p53 expression was found in 86/190 (45%) cancers, more commonly in serous carcinomas (77 vs 18% in high-grade endometrioid carcinomas, P<0.0001). BAF250a loss was associated with mismatch repair loss (P<0.0001) and normal p53 expression (P<0.0001). These associations were maintained in the subset analysis within the high-grade endometrioid (P=0.026 and P=0.0083, respectively) and serous carcinoma cases (P=0.0031 and P<0.0001, respectively). Survival analysis revealed a superior progression-free survival (P=0.017) for patients with BAF250a loss within the entire cohort but not within the high-grade endometrioid and serous subtypes. Additionally, data from The Cancer Genome Atlas were extracted to correlate mutations in ARID1A, TP53, and MMR genes; we found that ARID1A mutations were negatively associated with TP53 mutations but were unrelated to mismatch repair gene mutations. In conclusion, BAF250a loss is more common in high-grade endometrioid carcinomas than in other high-grade endometrial cancers and is associated with mismatch repair deficiency and normal p53 expression.


Subject(s)
DNA Mismatch Repair/genetics , Endometrial Neoplasms/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Aged , DNA Mutational Analysis , DNA-Binding Proteins , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Nuclear Proteins/genetics , Transcription Factors/genetics
11.
BMC Cancer ; 14: 120, 2014 Feb 22.
Article in English | MEDLINE | ID: mdl-24559118

ABSTRACT

BACKGROUND: Ovarian cancer is now recognized as a number of distinct diseases primarily defined by histological subtype. Both clear cell ovarian carcinomas (CCC) and ovarian endometrioid carcinomas (EC) may arise from endometriosis and frequently harbor mutations in the ARID1A tumor suppressor gene. We studied the influence of histological subtype on protein expression with reverse phase protein array (RPPA) and assessed proteomic changes associated with ARID1A mutation/BAF250a expression in EC and CCC. METHODS: Immunohistochemistry (IHC) for BAF250a expression was performed on 127 chemotherapy-naive ovarian carcinomas (33 CCC, 29 EC, and 65 high-grade serous ovarian carcinomas (HGSC)). Whole tumor lysates were prepared from frozen banked tumor samples and profiled by RPPA using 116 antibodies. ARID1A mutations were identified by exome sequencing, and PIK3CA mutations were characterized by MALDI-TOF mass spectrometry. SAM (Significance Analysis of Microarrays) was performed to determine differential protein expression by histological subtype and ARID1A mutation status. Multivariate logistic regression was used to assess the impact of ARID1A mutation status/BAF250a expression on AKT phosphorylation (pAKT). PIK3CA mutation type and PTEN expression were included in the model. BAF250a knockdown was performed in 3 clear cell lines using siRNA to ARID1A. RESULTS: Marked differences in protein expression were observed that are driven by histotype. Compared to HGSC, SAM identified over 50 proteins that are differentially expressed in CCC and EC. These included PI3K/AKT pathway proteins, those regulating the cell cycle, apoptosis, transcription, and other signaling pathways including steroid hormone signaling. Multivariate models showed that tumors with loss of BAF250a expression showed significantly higher levels of AKT-Thr308 and AKT-Ser473 phosphorylation (p < 0.05). In 31 CCC cases, pAKT was similarly significantly increased in tumors with BAF250a loss on IHC. Knockdown of BAF250a by siRNA in three CCC cell lines wild type for ARID1A showed no increase in either pAKT-Thr308 or pAKT-S473 suggesting that pAKT in tumor tissues is indirectly regulated by BAF250a expression. CONCLUSIONS: Proteomic assessment of CCC and EC demonstrates remarkable differences in protein expression that are dependent on histotype, thereby further characterizing these cancers. AKT phosphorylation is associated with ARID1A/BAF250a deficient tumors, however in ovarian cancers the mechanism remains to be elucidated.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/metabolism , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/metabolism , Mutation , Proteome , Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/pathology , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/pathology , Class I Phosphatidylinositol 3-Kinases , Cluster Analysis , DNA Mutational Analysis , DNA-Binding Proteins , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Neoplasm Grading , Neoplasm Staging , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Patient Outcome Assessment , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Prognosis , Proteomics/methods , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Transcription Factors/genetics
12.
Int J Gynecol Pathol ; 33(6): 560-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25272294

ABSTRACT

The contemporary oncologic pathology report conveys diagnostic, prognostic, predictive, and hereditary predisposition information. Each component may be premised on a morphologic feature or a biomarker. Clinical validity and reproducibility are paramount as is standardization of reporting and clinical response to ensure individualization of patient care. Regarding hereditary predisposition, morphology-based genetic referral systems in some instances have eclipsed genealogy-based systems, for example, cell type in ovarian cancer and BRCA screening. In other instances such as Lynch syndrome, morphology-based schemas supplement clinical schemas and there is an emerging standard of care for reflex biomarker testing. Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome predisposes patients to uterine and cutaneous leiomyomas (LMs) and renal cell carcinomas (RCCs). Several authors have emphasized the role pathologists may play in identifying this syndrome by recognizing the morphologic characteristics of syndromic uterine LMs and RCCs. Recently immunohistochemical overexpression of S-(2-succinyl) cysteine (2SC) has been demonstrated as a robust biomarker of mutation status in tumors from HLRCC patients. In this blinded control-cohort study we demonstrate that the proposed morphologic criteria used to identify uterine LMs in HLRCC syndrome are largely irreproducible among pathologists and lack sufficient robustness to serve as a trigger to triage cases for 2SC immunohistochemistry or patients for further family/personal history inquiry. Although refinement of morphologic criteria can be considered, in view of the availability of a clinically robust biomarker, consideration should be given to reflex testing of uterine LMs with an appropriate age cut off or in the setting of a suspicious family history.


Subject(s)
Leiomyomatosis/diagnosis , Pathology, Clinical/standards , Skin Neoplasms/diagnosis , Uterine Neoplasms/diagnosis , Adult , Aged , Cohort Studies , Female , Humans , Immunohistochemistry , Middle Aged , Neoplastic Syndromes, Hereditary , Reproducibility of Results , Sensitivity and Specificity
13.
J Pathol ; 231(1): 21-34, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23780408

ABSTRACT

High-grade serous ovarian cancer (HGSC) is characterized by poor outcome, often attributed to the emergence of treatment-resistant subclones. We sought to measure the degree of genomic diversity within primary, untreated HGSCs to examine the natural state of tumour evolution prior to therapy. We performed exome sequencing, copy number analysis, targeted amplicon deep sequencing and gene expression profiling on 31 spatially and temporally separated HGSC tumour specimens (six patients), including ovarian masses, distant metastases and fallopian tube lesions. We found widespread intratumoural variation in mutation, copy number and gene expression profiles, with key driver alterations in genes present in only a subset of samples (eg PIK3CA, CTNNB1, NF1). On average, only 51.5% of mutations were present in every sample of a given case (range 10.2-91.4%), with TP53 as the only somatic mutation consistently present in all samples. Complex segmental aneuploidies, such as whole-genome doubling, were present in a subset of samples from the same individual, with divergent copy number changes segregating independently of point mutation acquisition. Reconstruction of evolutionary histories showed one patient with mixed HGSC and endometrioid histology, with common aetiologic origin in the fallopian tube and subsequent selection of different driver mutations in the histologically distinct samples. In this patient, we observed mixed cell populations in the early fallopian tube lesion, indicating that diversity arises at early stages of tumourigenesis. Our results revealed that HGSCs exhibit highly individual evolutionary trajectories and diverse genomic tapestries prior to therapy, exposing an essential biological characteristic to inform future design of personalized therapeutic solutions and investigation of drug-resistance mechanisms.


Subject(s)
Cystadenocarcinoma, Serous/genetics , DNA Mutational Analysis/methods , Gene Expression Regulation, Neoplastic , Genetic Variation/genetics , Ovarian Neoplasms/genetics , Aged , Clone Cells , Cystadenocarcinoma, Serous/secondary , Disease Progression , Drug Resistance , Female , Gene Dosage , Gene Expression Profiling , Humans , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/pathology , Real-Time Polymerase Chain Reaction
14.
J Pathol ; 229(1): 111-20, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22899400

ABSTRACT

Mucinous ovarian carcinomas (MCs) typically do not respond to current conventional therapy. We have previously demonstrated amplification of HER2 in 6 of 33 (18.2%) mucinous ovarian carcinomas (MCs) and presented anecdotal evidence of response with HER2-targeted treatment in a small series of women with recurrent HER2-amplified (HER2+) MC. Here, we explore HER2 amplification and KRAS mutation status in an independent cohort of 189 MCs and 199 mucinous borderline ovarian tumours (MBOTs) and their association to clinicopathological features. HER2 status was assessed by immunohistochemistry (IHC), FISH, and CISH, and interpreted per ASCO/CAP guidelines, with intratumoural heterogeneity assessment on full sections, where available. KRAS mutation testing was performed with Sanger sequencing. Stage and grade were associated with recurrence on both univariate and multivariate analysis (p < 0.001). Assessment of HER2 status revealed overexpression/amplification of HER2 in 29/154 (18.8%) MCs and 11/176 (6.2%) MBOTs. There was excellent agreement between IHC, FISH, and CISH assessment of HER2 status (perfect concordance of HER2 0 or 1+ IHC with non-amplified status, and 3+ IHC with amplified status). KRAS mutations were seen in 31/71 (43.6%) MCs and 26/33 (78.8%) MBOTs, and were near mutually exclusive of HER2 amplification. In the 189 MC cases, a total of 54 recurrences and 59 deaths (53 of progressive disease) were observed. Within MCs, either HER2 amplification/overexpression or KRAS mutation was associated with decreased likelihood of disease recurrence (p = 0.019) or death (p = 0.0041) when compared to cases with neither feature. Intratumoural heterogeneity was noted in 26% of HER2-overexpressing cases. These data support the stratification of MCs for the testing of new treatments, with HER2-targeted therapy as a viable option for HER2+ advanced or recurrent disease. Further research is required to delineate the molecular and clinical features of the ∼34% of MC cases with neither HER2 amplification nor KRAS mutations.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Biomarkers, Tumor/genetics , Gene Amplification , Ovarian Neoplasms/genetics , Receptor, ErbB-2/genetics , Adenocarcinoma, Mucinous/chemistry , Adenocarcinoma, Mucinous/drug therapy , Adenocarcinoma, Mucinous/mortality , Adenocarcinoma, Mucinous/secondary , Adolescent , Adult , Aged , Aged, 80 and over , Australia , Biomarkers, Tumor/analysis , Canada , DNA Mutational Analysis , Disease Progression , Disease-Free Survival , Female , Genetic Predisposition to Disease , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Middle Aged , Molecular Targeted Therapy , Multivariate Analysis , Mutation , Neoplasm Grading , Neoplasm Recurrence, Local , Neoplasm Staging , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Phenotype , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Receptor, ErbB-2/analysis , Risk Assessment , Risk Factors , Time Factors , United States , Young Adult , ras Proteins/genetics
15.
Lung ; 192(4): 467-72, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24792232

ABSTRACT

BACKGROUND: An imbalance between proteolytic enzymes and their inhibitors is thought to be involved in the pathogenesis of chronic obstructive pulmonary disease. Matrix metalloproteinase-1, also known as interstitial collagenase, has been implicated as a potentially important proteinase in the genesis of chronic obstructive pulmonary disease and, more specifically, emphysema. METHODS: We performed quantitative immunohistochemical assessment of matrix metalloproteinase-1 expression in the resected lung of 20 smokers/ex-smokers who had varying severity of airflow obstruction and emphysema and compared this with the lungs of 5 nonsmokers. Emphysema was measured using a morphometric measure of the lungs' surface area/volume ratio and with qualitative and quantitative computed tomography (CT) measures of emphysema. RESULTS: There were significantly more matrix metalloproteinase-1-expressing alveolar macrophages and type II pneumocytes as well as a greater percentage of small airways that stained positively for matrix metalloproteinase-1 in the lungs of smokers than in those of nonsmokers (p < 0.0001, p < 0.0001, and p = 0.0003, respectively). The extent of staining of type II pneumocytes and airways for matrix metalloproteinase-1 was significantly related to the extent of smoking (p = 0.012 and p = 0.013, respectively). In addition, the extent of matrix metalloproteinase-1 staining of alveolar macrophages was related to the lung surface area/volume ratio and to qualitative estimates of emphysema on CT. CONCLUSION: These findings suggest that cigarette smoking increases expression of matrix metalloproteinase-1 in alveolar macrophages as well as in alveolar and small airway epithelial cells. Smokers who develop emphysema have increased alveolar macrophage expression of matrix metalloproteinase-1.


Subject(s)
Alveolar Epithelial Cells/enzymology , Lung/enzymology , Macrophages, Alveolar/enzymology , Matrix Metalloproteinase 1/analysis , Pulmonary Emphysema/enzymology , Smoking/metabolism , Adult , Aged , Case-Control Studies , Female , Humans , Immunohistochemistry , Lung/diagnostic imaging , Lung/physiopathology , Male , Middle Aged , Pulmonary Emphysema/diagnostic imaging , Pulmonary Emphysema/etiology , Pulmonary Emphysema/physiopathology , Respiratory Function Tests , Severity of Illness Index , Smoking/adverse effects , Smoking/physiopathology , Tomography, X-Ray Computed , Up-Regulation
16.
J Obstet Gynaecol Can ; 36(8): 708-715, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25222166

ABSTRACT

OBJECTIVE: Ovarian cancer treatments and outcomes vary substantially, yielding a diverse group of survivors. Few data exist on quality of life (QoL) concerns and the foremost needs of these patients. Our goal was to conduct a pilot study to determine the QoL needs of ovarian cancer survivors to establish priorities for future interventions. METHODS: In this cross-sectional study, the European Organization for Research and Treatment of Cancer Quality of Life Questionnaires (EORTC QLQ-C30 and OV28) QoL questionnaires and one investigator-derived questionnaire were administered in an outpatient setting. Clinical parameters were abstracted and tested for associations with QoL measures. RESULTS: A total of 102 women consented to participate and completed all components. Their mean age was 58 years (range 29 to 85), with 80% having epithelial ovarian carcinoma and 66% high-grade serous carcinoma. Women with stage I (28%), II (15%), III (47%), and IV (10%) lesions were represented in the primary treatment (25%), surveillance (46%), recurrent (23%), and palliative (7%) phases of the survivorship continuum. Fifty-one percent characterized their disease burden as "quite a bit" or "very much," and this did not vary by histology or diagnoses. Global QoL did not vary by clinico-pathologic parameters. Cardiovascular and respiratory comorbidities were associated with EORTC scores in physical functioning (P=0.027 for cardiovascular and P=0.041 for respiratory), global QoL (P=0.03 for cardiovascular and P=0.039 for respiratory), and sexual health (P=0.025 for cardiovascular). Task completion/memory/concentration, anxiety, and fatigue were the distress categories given highest priority by respondents. CONCLUSION: In women with ovarian cancer, clinical factors such as age, stage, and histology did not have a significant impact on QoL. Psychosocial factors have a larger impact on global QoL than physical symptoms.


Objectif : Les traitements contre le cancer de l'ovaire et leurs résultats varient considérablement, il en résulte donc un groupe diversifié de survivantes. Nous ne disposons que de peu de données sur les questions liées à la qualité de vie (QdV) de ces patientes et sur leurs besoins les plus criants. Nous avions pour objectif de mener une étude pilote visant à déterminer les besoins des survivantes du cancer de l'ovaire en matière de QdV afin d'établir les priorités pour ce qui est des futures interventions. Méthodes : Dans le cadre de cette étude transversale, les questionnaires sur la QdV de la European Organization for Research and Treatment of Cancer Quality of Life Questionnaires (EORTC QLQ-C30 et OV28) et un questionnaire formulé par les chercheurs ont été administrés au sein d'une clinique externe. Les paramètres cliniques ont été résumés et analysés en vue d'y déceler des associations avec les mesures de la QdV. Résultats : Au total, 102 femmes ont consenti à participer à l'étude et ont rempli toutes les composantes requises. Leur âge moyen était de 58 ans (plage : de 29 à 85); 80 % d'entre elles présentaient un carcinome épithélial de l'ovaire et 66 % présentaient un carcinome séreux de haut grade histologique. Les femmes présentant des lésions de stade I (28 %), de stade II (15 %), de stade III (47 %) et de stade IV (10 %) étaient représentées dans les phases « traitement primaire ¼ (25 %), « surveillance ¼ (46 %), « récurrent ¼ (23 %) et « palliatif ¼ (7 %) du continuum de la survie. Cinquante et un pour cent des répondantes ont caractérisé le fardeau de la maladie comme étant « plutôt lourd ¼ ou « très lourd ¼ et cette façon de répondre ne variait pas en fonction de l'histologie ou du diagnostic. La QdV globale ne variait pas en fonction des paramètres clinico-pathologiques. Des comorbidités cardiovasculaires et respiratoires ont été associées aux scores EORTC en ce qui concerne le fonctionnement physique (P = 0,027 pour ce qui est des comorbidités cardiovasculaires et P = 0,041 pour ce qui est des comorbidités respiratoires), la QdV globale (P = 0,03 pour ce qui est des comorbidités cardiovasculaires et P = 0,039 pour ce qui est des comorbidités respiratoires) et la santé sexuelle (P = 0,025 pour ce qui est des comorbidités cardiovasculaires). L'incapacité d'achever une tâche / les troubles de la mémoire et de la concentration, l'anxiété et la fatigue figuraient parmi les catégories de détresse auxquelles les participantes ont accordé la priorité absolue. Conclusion : Chez les femmes qui présentent un cancer de l'ovaire, des facteurs cliniques tels que l'âge, le stade et l'histologie n'exerçaient pas un effet significatif sur la QdV. Les facteurs psychosociaux exercent un effet plus important sur la QdV globale que les symptômes physiques.


Subject(s)
Ovarian Neoplasms/psychology , Quality of Life/psychology , Survivors/psychology , Adult , Aged , Aged, 80 and over , Analysis of Variance , Cross-Sectional Studies , Female , Humans , Middle Aged , Pilot Projects
17.
Cancer Res Commun ; 4(6): 1561-1565, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38837892

ABSTRACT

Cancer-related fatigue (CRF) continues to be a challenging phenomenon that is often under-reported and poorly understood. With etiologies in both disease and treatment manifesting as a symptom and a side effect respectively, CRF is highly incident and presents a significant clinical problem that impacts survivorship. We conducted a survey to ascertain the patient reported incidence of symptoms and side effects for people with lymphoma or chronic lymphocytic leukemia. We found that CRF was enhanced in those who received more intense therapies that coincided with more aggressive lymphoma subtypes. These data illuminate an unmet need among patients with lymphoma and provides an opportunity to further refine treatment regimens to reduce the burden of CRF in this vulnerable population. SIGNIFICANCE: CRF is a highly incident phenomenon in lymphoma that can be ascribed to a combination of causes. We have demonstrated substantial variability across various subtypes of lymphoma and have estimated that nearly half of the reported fatigue comes from treatment. Increased screening for and monitoring of fatigue will yield favorable health-related quality of life that will benefit health technology assessment activities and yield improved outcomes for patients.


Subject(s)
Fatigue , Lymphoma , Quality of Life , Humans , Fatigue/etiology , Fatigue/epidemiology , Lymphoma/epidemiology , Lymphoma/complications , Male , Female , Middle Aged , Aged , Surveys and Questionnaires , Adult , Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology , Leukemia, Lymphocytic, Chronic, B-Cell/complications , Incidence
18.
Cancer Metab ; 12(1): 28, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39363341

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease characterized by complex metabolic rewiring that enables growth in changing nutrient availability and oxygen conditions. Transcriptome-based prognostic PDAC tumor subtypes, known as 'basal-like' and 'classical' subtypes are associated with differences in metabolic gene expression including genes involved in glycolysis. Tumor subtype-specific metabolism phenotypes may provide new targets for treatment development in PDAC, but their functional relevance has not been fully elucidated. We aimed to investigate differences in metabolic profiles and transcriptomes in tumor models derived from patients with basal-like and classical tumors. METHODS: Patient-derived organoids (PDOs) were established from tumor biopsies collected from patients with metastatic PDAC, including three PDOs from basal-like and five PDOs from classical tumors. Metabolic analyses included assessment of differences in metabolic activity using Seahorse Glycolysis and Mito Stress tests and 13C-glucose metabolites tracing analysis. In order to investigate the influence of mitochondrial pyruvate transport on metabolic differences, PDOs were treated with the mitochondrial pyruvate carrier 1 (MPC1) inhibitor UK-5099. Prognostic relevance of MPC1 was determined using a tumor tissue microarray (TMA) in resectable, and proteomics profiling in metastatic PDAC datasets. Whole genome and transcriptome sequencing, differential gene expression and gene set enrichment analyses were performed in PDOs. RESULTS: Metastatic PDAC PDOs showed subtype-specific differences in glycolysis and oxidative phosphorylation (OXPHOS). Basal-like tumor-derived PDOs had a lower baseline extracellular acidification rate, but higher glycolytic reserves and oxygen consumption rate (OCR) than classical tumor-derived PDOs. OCR difference was eliminated following treatment with UK-5099. In the 13C-glucose metabolites tracing experiment, a basal-like tumor PDO showed lower fractions of some M + 2 metabolites but higher sensitivity to UK-5099 mediated reduction in M + 2 metabolites than a classical tumor PDO. Protein level analyses revealed lower MPC1 protein levels in basal-like PDAC cases and association of low MPC1 levels with clinicopathologic parameters of tumor aggressiveness in PDAC. PDO differential gene expression analyses identified additional subtype-specific cellular pathways and potential disease outcome biomarkers. CONCLUSIONS: Our findings point to distinct metabolic profiles in PDAC subtypes with basal-like tumor PDOs showing higher OXPHOS and sensitivity to MPC1 inhibition. Subtypes-specific metabolic vulnerabilities may be exploited for selective therapeutic targeting.

19.
N Engl J Med ; 363(16): 1532-43, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20942669

ABSTRACT

BACKGROUND: Ovarian clear-cell and endometrioid carcinomas may arise from endometriosis, but the molecular events involved in this transformation have not been described. METHODS: We sequenced the whole transcriptomes of 18 ovarian clear-cell carcinomas and 1 ovarian clear-cell carcinoma cell line and found somatic mutations in ARID1A (the AT-rich interactive domain 1A [SWI-like] gene) in 6 of the samples. ARID1A encodes BAF250a, a key component of the SWI­SNF chromatin remodeling complex. We sequenced ARID1A in an additional 210 ovarian carcinomas and a second ovarian clear-cell carcinoma cell line and measured BAF250a expression by means of immunohistochemical analysis in an additional 455 ovarian carcinomas. RESULTS: ARID1A mutations were seen in 55 of 119 ovarian clear-cell carcinomas (46%), 10 of 33 endometrioid carcinomas (30%), and none of the 76 high-grade serous ovarian carcinomas. Seventeen carcinomas had two somatic mutations each. Loss of the BAF250a protein correlated strongly with the ovarian clear-cell carcinoma and endometrioid carcinoma subtypes and the presence of ARID1A mutations. In two patients, ARID1A mutations and loss of BAF250a expression were evident in the tumor and contiguous atypical endometriosis but not in distant endometriotic lesions. CONCLUSIONS: These data implicate ARID1A as a tumor-suppressor gene frequently disrupted in ovarian clear-cell and endometrioid carcinomas. Since ARID1A mutation and loss of BAF250a can be seen in the preneoplastic lesions, we speculate that this is an early event in the transformation of endometriosis into cancer. (Funded by the British Columbia Cancer Foundation and the Vancouver General Hospital­University of British Columbia Hospital Foundation.).


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Carcinoma, Endometrioid/genetics , Endometriosis/complications , Mutation , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Transcription Factors/genetics , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Cell Line, Tumor , DNA-Binding Proteins , Endometriosis/pathology , Female , Gene Expression , Gene Expression Profiling , Genes, Tumor Suppressor , Humans , Nuclear Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Sequence Analysis, RNA , Transcription Factors/metabolism
20.
Mod Pathol ; 26(12): 1594-604, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23807777

ABSTRACT

Subclassification of endometrial carcinoma according to histological type shows variable interobserver agreement. The aim of this study was to assess specifically the interobserver agreement of histological type in high-grade endometrial carcinomas, recorded by gynecological pathologists from five academic centers across Canada. In a secondary aim, the agreement of consensus diagnosis with immunohistochemical marker combinations was assessed including six routine (TP53, CDKN2A (p16), ER, PGR, Ki67, and VIM) and six experimental immunohistochemical markers (PTEN, ARID1A, CTNNB1, IGF2BP3, HNF1B, and TFF3). The paired interobserver agreement ranged from κ 0.50 to 0.63 (median 0.58) and the intraobserver agreement from κ 0.49 to 0.67 (median 0.61). Consensus about histological type based on morphological assessment was reached in 72% of high-grade endometrial carcinomas. A seven-marker immunohistochemical panel differentiated FIGO grade 3 endometrioid from serous carcinoma with a 100% concordance rate compared with the consensus diagnosis. More practically, a three-marker panel including TP53, ER, and CDKN2A (p16) can aid in the differential diagnosis of FIGO grade 3 endometrioid from endometrial serous carcinoma. Our study demonstrates that the inter- and intraobserver reproducibility of histological type based on morphology alone are mostly moderate. Ancillary techniques such as immunohistochemical marker panels are likely needed to improve diagnostic reproducibility of histological types within high-grade endometrial carcinomas.


Subject(s)
Carcinoma, Endometrioid/pathology , Endometrial Neoplasms/pathology , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Cystadenocarcinoma, Serous/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Observer Variation , Reproducibility of Results , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL