Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Eur J Cancer ; 152: 78-89, 2021 07.
Article in English | MEDLINE | ID: mdl-34090143

ABSTRACT

AIM: The aim of the study was to assess the prognostic performance of a 6-gene molecular score (OncoMasTR Molecular Score [OMm]) and a composite risk score (OncoMasTR Risk Score [OM]) and to conduct a within-patient comparison against four routinely used molecular and clinicopathological risk assessment tools: Oncotype DX Recurrence Score, Ki67, Nottingham Prognostic Index and Clinical Risk Category, based on the modified Adjuvant! Online definition and three risk factors: patient age, tumour size and grade. METHODS: Biospecimens and clinicopathological information for 404 Irish women also previously enrolled in the Trial Assigning Individualized Options for Treatment [Rx] were provided by 11 participating hospitals, as the primary objective of an independent translational study. Gene expression measured via RT-qPCR was used to calculate OMm and OM. The prognostic value for distant recurrence-free survival (DRFS) and invasive disease-free survival (IDFS) was assessed using Cox proportional hazards models and Kaplan-Meier analysis. All statistical tests were two-sided ones. RESULTS: OMm and OM (both with likelihood ratio statistic [LRS] P < 0.001; C indexes = 0.84 and 0.85, respectively) were more prognostic for DRFS and provided significant additional prognostic information to all other assessment tools/factors assessed (all LRS P ≤ 0.002). In addition, the OM correctly classified more patients with distant recurrences (DRs) into the high-risk category than other risk classification tools. Similar results were observed for IDFS. DISCUSSION: Both OncoMasTR scores were significantly prognostic for DRFS and IDFS and provided additional prognostic information to the molecular and clinicopathological risk factors/tools assessed. OM was also the most accurate risk classification tool for identifying DR. A concise 6-gene signature with superior risk stratification was shown to increase prognosis reliability, which may help clinicians optimise treatment decisions.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/genetics , Breast Neoplasms/mortality , Breast/pathology , Neoplasm Recurrence, Local/epidemiology , Adult , Aged , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Disease-Free Survival , Female , Gene Expression Profiling , Genetic Testing/methods , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Observational Studies as Topic , Prognosis , Prospective Studies , Receptor, ErbB-2/analysis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/analysis , Receptors, Estrogen/metabolism , Receptors, Progesterone/analysis , Receptors, Progesterone/metabolism , Reproducibility of Results , Risk Assessment/methods , Risk Assessment/statistics & numerical data , Young Adult
2.
Expert Rev Mol Diagn ; 20(10): 1027-1037, 2020 10.
Article in English | MEDLINE | ID: mdl-32510287

ABSTRACT

INTRODUCTION: Tissue-based imaging has emerged as a critical tool in translational cancer research and is rapidly gaining traction within a clinical context. Significant progress has been made in the digital pathology arena, particularly in respect of brightfield and fluorescent imaging. Critically, the cellular context of molecular alterations occurring at DNA, RNA, or protein level within tumor tissue is now being more fully appreciated. Moreover, the emergence of novel multi-marker imaging approaches can now provide unprecedented insights into the tumor microenvironment, including the potential interplay between various cell types. AREAS COVERED: This review summarizes the recent developments within the field of tissue-based imaging, centering on the application of these approaches in oncology research and clinical practice. EXPERT OPINION: Significant advances have been made in digital pathology during the last 10 years. These include the use of quantitative image analysis algorithms, predictive artificial intelligence (AI) on large datasets of H&E images, and quantification of fluorescence multiplexed tissue imaging data. We believe that new methodologies that can integrate AI-derived histologic data with omic data, together with other forms of imaging data (such as radiologic image data), will enhance our ability to deliver better diagnostics and treatment decisions to the cancer patient.


Subject(s)
Biomarkers, Tumor , Molecular Imaging/methods , Neoplasms/pathology , Artificial Intelligence , Disease Management , Fluorescent Antibody Technique/methods , Fluorescent Antibody Technique/standards , Image Processing, Computer-Assisted , Immunohistochemistry/methods , Immunohistochemistry/standards , Medical Oncology/methods , Medical Oncology/standards , Molecular Imaging/standards , Neoplasms/diagnostic imaging , Neoplasms/etiology , Practice Patterns, Physicians' , Translational Research, Biomedical , Tumor Microenvironment
3.
Cancer Res ; 77(9): 2186-2190, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28428271

ABSTRACT

Reverse engineering of transcriptional networks using gene expression data enables identification of genes that underpin the development and progression of different cancers. Methods to this end have been available for over a decade and, with a critical mass of transcriptomic data in the oncology arena having been reached, they are ever more applicable. Extensive and complex networks can be distilled into a small set of key master transcriptional regulators (MTR), genes that are very highly connected and have been shown to be involved in processes of known importance in disease. Interpreting and validating the results of standardized bioinformatic methods is of crucial importance in determining the inherent value of MTRs. In this review, we briefly describe how MTRs are identified and focus on providing an overview of how MTRs can and have been validated for use in clinical decision making in malignant diseases, along with serving as tractable therapeutic targets. Cancer Res; 77(9); 2186-90. ©2017 AACR.


Subject(s)
Gene Regulatory Networks/genetics , Neoplasms/genetics , Transcription, Genetic , Transcriptome/genetics , Computational Biology , Gene Expression Regulation, Neoplastic/genetics , Humans
4.
FEBS J ; 282(18): 3455-73, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26094870

ABSTRACT

The majority of women diagnosed with lymph node-negative breast cancer are unnecessarily treated with damaging chemotherapeutics after surgical resection. This highlights the importance of understanding and more accurately predicting patient prognosis. In the present study, we define the transcriptional networks regulating well-established prognostic gene expression signatures. We find that the same set of transcriptional regulators consistently lie upstream of both 'prognosis' and 'proliferation' gene signatures, suggesting that a central transcriptional network underpins a shared phenotype within these signatures. Strikingly, the master transcriptional regulators within this network predict recurrence risk for lymph node-negative breast cancer better than currently used multigene prognostic assays, particularly in estrogen receptor-positive patients. Simultaneous examination of p16(INK4A) expression, which predicts tumours that have bypassed cellular senescence, revealed that intermediate levels of p16(INK4A) correlate with an intact pRB pathway and improved survival. A combination of these master transcriptional regulators and p16(INK4A), termed the OncoMasTR score, stratifies tumours based on their proliferative and senescence capacity, facilitating a clearer delineation of lymph node-negative breast cancer patients at high risk of recurrence, and thus requiring chemotherapy. Furthermore, OncoMasTR accurately classifies over 60% of patients as 'low risk', an improvement on existing prognostic assays, which has the potential to reduce overtreatment in early-stage patients. Taken together, the present study provides new insights into the transcriptional regulation of cellular proliferation in breast cancer and provides an opportunity to enhance and streamline methods of predicting breast cancer prognosis.


Subject(s)
Breast Neoplasms/genetics , Gene Regulatory Networks , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Proliferation/genetics , Cells, Cultured , Cellular Senescence/genetics , Cohort Studies , Female , Genes, p16 , Humans , Lymphatic Metastasis/genetics , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Middle Aged , Prognosis , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Estrogen/metabolism , Risk Factors , Tissue Array Analysis
5.
Oncotarget ; 6(14): 12209-23, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25906750

ABSTRACT

FK506-binding protein-like (FKBPL) has established roles as an anti-tumor protein, with a therapeutic peptide based on this protein, ALM201, shortly entering phase I/II clinical trials. Here, we evaluated FKBPL's prognostic ability in primary breast cancer tissue, represented on tissue microarrays (TMA) from 3277 women recruited into five independent retrospective studies, using immunohistochemistry (IHC). In a meta-analysis, FKBPL levels were a significant predictor of BCSS; low FKBPL levels indicated poorer breast cancer specific survival (BCSS) (hazard ratio (HR) = 1.30, 95% confidence interval (CI) 1.14-1.49, p < 0.001). The prognostic impact of FKBPL remained significant after adjusting for other known prognostic factors (HR = 1.25, 95% CI 1.07-1.45, p = 0.004). For the sub-groups of 2365 estrogen receptor (ER) positive patients and 1649 tamoxifen treated patients, FKBPL was significantly associated with BCSS (HR = 1.34, 95% CI 1.13-1.58, p < 0.001, and HR = 1.25, 95% CI 1.04-1.49, p = 0.02, respectively). A univariate analysis revealed that FKBPL was also a significant predictor of relapse free interval (RFI) within the ER positive patient group, but it was only borderline significant within the smaller tamoxifen treated patient group (HR = 1.32 95% CI 1.05-1.65, p = 0.02 and HR = 1.23 95% CI 0.99-1.54, p = 0.06, respectively). The data suggests a role for FKBPL as a prognostic factor for BCSS, with the potential to be routinely evaluated within the clinic.


Subject(s)
Breast Neoplasms/genetics , Immunophilins/genetics , Immunophilins/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cohort Studies , Female , Humans , Precision Medicine , Prognosis , Survival Analysis , Tacrolimus Binding Proteins
6.
Biochem Soc Trans ; 39(2): 663-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21428958

ABSTRACT

FKBPs (FK506-binding proteins) have long been recognized as key regulators of the response to immunosuppressant drugs and as co-chaperones of steroid receptor complexes. More recently, evidence has emerged suggesting that this diverse protein family may also represent cancer biomarkers owing to their roles in cancer progression and response to treatment. FKBPL (FKBP-like) is a novel FKBP with roles in GR (glucocorticoid receptor), AR (androgen receptor) and ER (oestrogen receptor) signalling. FKBPL binds Hsp90 (heat-shock protein 90) and modulates translocation, transcriptional activation and phosphorylation of these steroid receptors. It has been proposed as a novel prognostic and predictive biomarker, where high levels predict for increased recurrence-free survival in breast cancer patients and enhanced sensitivity to endocrine therapy. Since this protein family has roles in a plethora of signalling pathways, its members represent novel prognostic markers and therapeutic targets for cancer diagnosis and treatment.


Subject(s)
Biomarkers, Tumor/physiology , Immunophilins/physiology , Neoplasms/diagnosis , Tacrolimus Binding Proteins/physiology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Immunophilins/genetics , Immunophilins/metabolism , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction/physiology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
7.
J Cell Sci ; 123(Pt 19): 3316-28, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20844151

ABSTRACT

Mammary epithelial cells organize in three dimensions and generate acini when supported on laminin-rich extracellular matrix. Acinus formation begins with the apicobasal polarisation of the outer cells of the assembly and the withdrawal of these cells from the cell cycle. Internal cells then clear out to form a hollow lumen. Here, we show that PKCζ is phosphorylated (at T410) and activated in the early stages of acinus formation in both primary cells and MCF10A cells, and during mammary tree maturation in vivo. Phospho-PKCζ colocalised with tight junction components and bound to the Par polarising complex in developing acini. To further investigate the importance of PKCζ phosphorylation in this context, acinus formation was studied in MCF10A cells overexpressing non-phosphorylatable (T410A) or 'constitutively phosphorylated' (T410E) PKCζ. In both cell types, acinus-associated cell polarisation and lumen clearance were compromised, emphasising the importance of regulated phosphorylation of PKCζ at T410 for successful acinus formation. PKCζ can be activated in a phosphorylation (at T410)-dependent and a phosphorylation-independent manner. Cells overexpressing a complete kinase-deficient PKCζ (K281W) displayed a cell polarising deficit, but also generated large 'multi-acinar' structures with associated early lumenal cell hyperproliferation. Therefore our data shows, for the first time, that two separable PKCζ activities (one phosphorylation-dependent, the other not) are required to support the cell polarisation and proliferation restriction that underpins successful acinus formation. Paralleling these contributions, we found that low levels of PKCζ mRNA expression are associated with more 'poorly differentiated' tumours and a poor outcome in a cohort of 295 breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Protein Kinase C/metabolism , Adult , Animals , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Polarity/genetics , Cell Transformation, Neoplastic , Disease Progression , Epithelial Cells/pathology , Female , Humans , Hyperplasia/genetics , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Mice , Mice, Inbred Strains , Microarray Analysis , Mutant Proteins/genetics , Organogenesis/genetics , Phosphorylation , Pregnancy , Protein Kinase C/genetics , Survival Analysis , Transgenes/genetics
8.
Breast Cancer Res ; 12(4): R59, 2010.
Article in English | MEDLINE | ID: mdl-20682066

ABSTRACT

INTRODUCTION: The homeobox-containing transcription factor muscle segment homeobox 2 (Msx2) plays an important role in mammary gland development. However, the clinical implications of Msx2 expression in breast cancer are unclear. The aims of this study were to investigate the potential clinical value of Msx2 as a breast cancer biomarker and to clarify its functional role in vitro. METHODS: Msx2 gene expression was first examined in a well-validated breast cancer transcriptomic dataset of 295 patients. Msx2 protein expression was then evaluated by immunohistochemistry in a tissue microarray (TMA) containing 281 invasive breast tumours. Finally, to assess the functional role of Msx2 in vitro, Msx2 was ectopically expressed in a highly invasive breast tumour cell line (MDA-MB-231) and an immortalised breast cell line (MCF10a), and these cell lines were examined for changes in growth rate, cell death and cell signalling. RESULTS: Examination of Msx2 mRNA expression in a breast cancer transcriptomic dataset demonstrated that increased levels of Msx2 were associated with good prognosis (P = 0.011). Evaluation of Msx2 protein expression on a TMA revealed that Msx2 was detectable in both tumour cell nuclei and cytoplasm. Cytoplasmic Msx2 expression was associated with low grade tumours (P = 0.012) and Ki67 negativity (P = 0.018). Nuclear Msx2 correlated with low-grade tumours (P = 0.015), estrogen receptor positivity (P = 0.038), low Ki67 (P = 0.005) and high cyclin D1 expression (P = 0.037). Increased cytoplasmic Msx2 expression was associated with a prolonged breast cancer-specific survival (P = 0.049), recurrence-free survival (P = 0.029) and overall survival (P = 0.019). Ectopic expression of Msx2 in breast cell lines resulted in radically decreased cell viability mediated by induction of cell death via apoptosis. Further analysis of Msx2-expressing cells revealed increased levels of p21 and phosphorylated extracellular signal-regulated kinase (ERK) and decreased levels of Survivin and the 'split ends' (SPEN) protein family member RBM15. CONCLUSIONS: We conclude that increased Msx2 expression results in improved outcome for breast cancer patients, possibly by increasing the likelihood of tumour cell death by apoptosis.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Homeodomain Proteins/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Immunohistochemistry/statistics & numerical data , Kaplan-Meier Estimate , Middle Aged , Prognosis , Proportional Hazards Models , Signal Transduction , Tissue Array Analysis
9.
Int J Cancer ; 124(9): 2088-97, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19142967

ABSTRACT

The role of intercellular tight junctions in breast epithelial cells is traditionally thought to be in maintaining polarity and barrier function. However, claudin-4, a tight junction protein, is overexpressed in breast tumour cells compared to normal epithelial cells, which generally corresponds to a loss in polarity. The aim of this study was to investigate the distribution and potential clinical value of claudin-4 in breast cancer, and to evaluate its usefulness as a prognostic and predictive biomarker. Expression of claudin-4 was initially examined by Western blot analysis in a cohort of 88 breast tumours, and was found to correlate positively with tumour grade and negatively with ER. Claudin-4 expression was then evaluated by immunohistochemistry in a larger cohort of 299 tumours represented on a tissue microarray. Claudin-4 expression correlated positively with tumour grade and Her2, and negatively with ER. High claudin-4 expression was also associated with worse breast cancer-specific survival (p = 0.003), recurrence-free survival (p = 0.025) and overall survival (p = 0.034). Multivariate analysis revealed that claudin-4 independently predicted survival in the entire cohort (HR 1.95; 95%CI 1.01-3.79; p = 0.047) and in the ER positive subgroup treated with adjuvant tamoxifen (HR 4.34; 95%CI 1.14-16.53; p = 0.032). This relationship between increased claudin-4 expression and adverse outcome was validated at the mRNA level in a DNA microarray dataset of 295 breast tumours. We conclude that high levels of claudin-4 protein are associated with adverse outcome in breast cancer patients, including the subgroup of patients treated with adjuvant tamoxifen.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/metabolism , Carcinoma, Medullary/metabolism , Membrane Proteins/metabolism , Adenocarcinoma, Mucinous/drug therapy , Adenocarcinoma, Mucinous/secondary , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/secondary , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/secondary , Carcinoma, Medullary/drug therapy , Carcinoma, Medullary/secondary , Claudin-4 , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Membrane Proteins/genetics , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , Receptor, ErbB-2/metabolism , Survival Rate , Tamoxifen/therapeutic use , Tissue Array Analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL