Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
EBioMedicine ; 88: 104436, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36708693

ABSTRACT

BACKGROUND: Protein markers of cellular proliferation, hypoxia, apoptosis, cell cycle checkpoints, growth factor signalling and inflammation in localised prostate tumours have previously shown prognostic ability. A translational substudy within the CHHiP trial of radiotherapy fractionation evaluated whether these could improve prediction of prognosis and assist treatment stratification following either conventional or hypofractionated radiotherapy. METHODS: Using case:control methodology, patients with biochemical or clinical failure after radiotherapy (BCR) were matched to patients without recurrence according to established prognostic factors (Gleason score, presenting PSA, tumour-stage) and fractionation schedule. Immunohistochemical (IHC) staining of diagnostic biopsy sections was performed and scored for HIF1α, Bcl-2, Ki67, Geminin, p16, p53, p-chk1 and PTEN. Univariable and multivariable conditional logistic regression models, adjusted for matching strata and age, estimated the prognostic value of each IHC biomarker, including interaction terms to determine BCR prediction according to fractionation. FINDINGS: IHC results were available for up to 336 tumours. PTEN, Geminin, mean Ki67 and max Ki67 were prognostic after adjusting for multiple comparisons and were fitted in a multivariable model (n = 212, 106 matched pairs). Here, PTEN and Geminin showed significant prediction of prognosis. No marker predicted BCR according to fractionation. INTERPRETATION: Geminin or Ki67, and PTEN, predicted response to radiotherapy independently of established prognostic factors. These results provide essential independent external validation of previous findings and confirm a role for these markers in treatment stratification. FUNDING: Cancer Research UK (BIDD) grant (A12518), Cancer Research UK (C8262/A7253), Department of Health, Prostate Cancer UK, Movember Foundation, NIHR Biomedical Research Centre at Royal Marsden/ICR.


Subject(s)
Prostatic Neoplasms , Male , Humans , Geminin , Ki-67 Antigen/metabolism , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/pathology , Dose Fractionation, Radiation
2.
Cell Death Differ ; 29(1): 54-64, 2022 01.
Article in English | MEDLINE | ID: mdl-34294889

ABSTRACT

Breast cancer heterogeneity has made it challenging to identify mechanisms critical to the initial stages of their genesis in vivo. Here, we sought to interrogate the role of YB-1 in newly arising human breast cancers as well as in established cell lines. In a first series of experiments, we found that short-hairpin RNA-mediated knockdown of YB-1 in MDA-MB-231 cells blocked both their local tumour-forming and lung-colonising activity in immunodeficient mice. Conversely, upregulated expression of YB-1 enhanced the poor in vivo tumorigenicity of T47D cells. We then found that YB-1 knockdown also inhibits the initial generation in mice of invasive ductal carcinomas and ductal carcinomas in situ from freshly isolated human mammary cells transduced, respectively, with KRASG12D or myristoylated-AKT1. Interestingly, increased expression of HIF1α and G3BP1, two YB-1 translational targets and elements of a stress-adaptive programme, mirrored the levels of YB-1 in both transformed primary and established MDA-MB-231 breast cancer cells.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Helicases/metabolism , Female , Humans , Mice , Poly-ADP-Ribose Binding Proteins , RNA Helicases/metabolism , RNA Recognition Motif Proteins , Transcription Factors/metabolism , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
3.
NPJ Breast Cancer ; 7(1): 19, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33649333

ABSTRACT

Despite increasing evidence supporting the clinical relevance of tumour infiltrating lymphocytes (TILs) in invasive breast cancer, TIL spatial variability within ductal carcinoma in situ (DCIS) samples and its association with progression are not well understood. To characterise tissue spatial architecture and the microenvironment of DCIS, we designed and validated a new deep learning pipeline, UNMaSk. Following automated detection of individual DCIS ducts using a new method IM-Net, we applied spatial tessellation to create virtual boundaries for each duct. To study local TIL infiltration for each duct, DRDIN was developed for mapping the distribution of TILs. In a dataset comprising grade 2-3 pure DCIS and DCIS adjacent to invasive cancer (adjacent DCIS), we found that pure DCIS cases had more TILs compared to adjacent DCIS. However, the colocalisation of TILs with DCIS ducts was significantly lower in pure DCIS compared to adjacent DCIS, which may suggest a more inflamed tissue ecology local to DCIS ducts in adjacent DCIS cases. Our study demonstrates that technological developments in deep convolutional neural networks and digital pathology can enable an automated morphological and microenvironmental analysis of DCIS, providing a new way to study differential immune ecology for individual ducts and identify new markers of progression.

4.
Stem Cell Reports ; 10(6): 1676-1686, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29874626

ABSTRACT

Human breast cancers referred to as "basal-like" are of interest because they lack effective therapies and their biology is poorly understood. The term basal-like derives from studies demonstrating tumor gene expression profiles that include some transcripts characteristic of the basal cells of the normal adult human mammary gland and others associated with a subset of normal luminal cells. Elucidating the mechanisms responsible for the profiles of basal-like tumors is an active area of investigation. More refined molecular analysis of patients' samples and genetic strategies to produce breast cancers de novo from defined populations of normal mouse mammary cells have served as complementary approaches to identify relevant pathway alterations. However, both also have limitations. Here, we review some of the underlying reasons, including the unifying concept that some normal luminal cells have both luminal and basal features, as well as some emerging new avenues of investigation.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Neoplasms, Basal Cell/etiology , Neoplasms, Basal Cell/metabolism , Animals , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Humans , Neoplasms, Basal Cell/pathology
5.
Int J Radiat Oncol Biol Phys ; 101(2): 309-315, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29559283

ABSTRACT

PURPOSE: To assess whether the cellular proliferation marker Ki67 provides prognostic information and predicts response to radiation therapy fractionation in patients with localized prostate tumors participating in a randomized trial of 3 radiation therapy fractionation schedules (74 Gy/37 fractions vs 60 Gy/20 fractions vs 57 Gy/19 fractions). METHODS AND MATERIALS: A matched case-control study design was used; patients with biochemical/clinical failure >2 years after radiation therapy (BCR) were matched 1:1 to patients without recurrence using established prognostic factors (Gleason score, prostate-specific antigen, tumor stage) and fractionation schedule. Immunohistochemistry was used to stain diagnostic biopsy specimens for Ki67, which were scored using the unweighted global method. Conditional logistic regression models estimated the prognostic value of mean and maximum Ki67 scores on BCR risk. Biomarker-fractionation interaction terms determined whether Ki67 was predictive of BCR by fractionation. RESULTS: Using 173 matched pairs, the median for mean and maximum Ki67 scores were 6.6% (interquartile range, 3.9%-9.8%) and 11.0% (interquartile range, 7.0%-15.0%) respectively. Both scores were significant predictors of BCR in models adjusted for established prognostic factors. Conditioning on matching variables and age, the odds of BCR were estimated to increase by 9% per 1% increase in mean Ki67 score (odds ratio 1.09; 95% confidence interval 1.04-1.15, P = .001). Interaction terms between Ki67 and fractionation schedules were not statistically significant. CONCLUSIONS: Diagnostic Ki67 did not predict BCR according to fractionation schedule in CHHiP; however, it was a strong independent prognostic factor for BCR.


Subject(s)
Dose Fractionation, Radiation , Ki-67 Antigen/analysis , Neoplasm Recurrence, Local , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/radiotherapy , Aged , Cell Proliferation , Humans , Immunohistochemistry , Logistic Models , Male , Matched-Pair Analysis , Neoplasm Grading , Odds Ratio , Predictive Value of Tests , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/pathology
6.
7.
Development ; 144(20): 3777-3788, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28870991

ABSTRACT

PTPRB is a transmembrane protein tyrosine phosphatase known to regulate blood vessel remodelling and angiogenesis. Here, we demonstrate that PTPRB negatively regulates branching morphogenesis in the mouse mammary epithelium. We show that Ptprb is highly expressed in adult mammary stem cells and also, although at lower levels, in oestrogen receptor-positive luminal cells. During mammary development, Ptprb expression is downregulated during puberty, a period of extensive ductal outgrowth and branching. In vivo shRNA knockdown of Ptprb in the cleared mammary fat pad transplant assay resulted in smaller epithelial outgrowths with an increased branching density and also increased branching in an in vitro organoid assay. Organoid branching was dependent on stimulation by FGF2, and Ptprb knockdown in mammary epithelial cells resulted in a higher level of fibroblast growth factor receptor (FGFR) activation and ERK1/2 phosphorylation, both at baseline and following FGF2 stimulation. Therefore, PTPRB regulates branching morphogenesis in the mammary epithelium by modulating the response of the FGFR signalling pathway to FGF stimulation. Considering the importance of branching morphogenesis in multiple taxa, our findings have general importance outside mammary developmental biology.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Mammary Glands, Animal/growth & development , Morphogenesis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Animals , Body Patterning , Epithelial Cells/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Mice , Neovascularization, Physiologic , Oligonucleotide Array Sequence Analysis , Organoids/growth & development , Phosphorylation , RNA, Small Interfering/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptors, Estrogen/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Stem Cells/cytology , Transgenes
8.
J Clin Oncol ; 33(25): 2772-9, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26215945

ABSTRACT

PURPOSE: To evaluate the relative effectiveness of letrozole compared with tamoxifen for patients with invasive ductal or lobular carcinoma. PATIENTS AND METHODS: Patients diagnosed with early-stage invasive ductal carcinoma (IDC) or classic invasive lobular carcinoma (ILC) who were randomly assigned onto the Breast International Group (BIG) 1-98 trial and who had centrally reviewed pathology data were included (N = 2,923). HER2-negative IDC and ILC were additionally classified as hormone receptor-positive with high (luminal B [LB] -like) or low (luminal A [LA] -like) proliferative activity by Ki-67 labeling index. Survival analyses were performed with weighted Cox models that used inverse probability of censoring weighted modeling. RESULTS: The median follow-up time was 8.1 years. In multivariable models for disease-free survival (DFS), significant interactions between treatment and histology (ILC or IDC; P = .006) and treatment and subgroup (LB like or LA like; P = .01) were observed. In the ILC subset, there was a 66% reduction in the hazard of a DFS event with letrozole for LB (hazard ratio [HR], 0.34; 95% CI, 0.21 to 0.55) and a 50% reduction for LA subtypes (HR, 0.50; 95% CI, 0.32 to 0.78). In the IDC subset, there was a significant 35% reduction in the hazard of a DFS event with letrozole for the LB subtype (HR, 0.65; 95% CI, 0.53 to 0.79), but no difference between treatments was noted for IDC and the LA subtype (HR, 0.95; 95% CI, 0.76 to 1.20). CONCLUSION: The magnitude of benefit of adjuvant letrozole is greater for patients diagnosed with lobular carcinoma versus ductal carcinoma.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Lobular/drug therapy , Nitriles/therapeutic use , Tamoxifen/therapeutic use , Triazoles/therapeutic use , Adult , Aromatase Inhibitors/therapeutic use , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Lobular/chemistry , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Ki-67 Antigen/analysis , Letrozole , Middle Aged , Receptor, ErbB-2/analysis , Treatment Outcome
9.
Dis Model Mech ; 7(5): 525-34, 2014 May.
Article in English | MEDLINE | ID: mdl-24626992

ABSTRACT

RUNX2, a master regulator of osteogenesis, is oncogenic in the lymphoid lineage; however, little is known about its role in epithelial cancers. Upregulation of RUNX2 in cell lines correlates with increased invasiveness and the capacity to form osteolytic disease in models of breast and prostate cancer. However, most studies have analysed the effects of this gene in a limited number of cell lines and its role in primary breast cancer has not been resolved. Using a human tumour tissue microarray, we show that high RUNX2 expression is significantly associated with oestrogen receptor (ER)/progesterone receptor (PR)/HER2-negative breast cancers and that patients with high RUNX2 expression have a poorer survival rate than those with negative or low expression. We confirm RUNX2 as a gene that has a potentially important functional role in triple-negative breast cancer. To investigate the role of this gene in breast cancer, we made a transgenic model in which Runx2 is specifically expressed in murine mammary epithelium under the control of the mouse mammary tumour virus (MMTV) promoter. We show that ectopic Runx2 perturbs normal development in pubertal and lactating animals, delaying ductal elongation and inhibiting lobular alveolar differentiation. We also show that the Runx2 transgene elicits age-related, pre-neoplastic changes in the mammary epithelium of older transgenic animals, suggesting that elevated RUNX2 expression renders such tissue more susceptible to oncogenic changes and providing further evidence that this gene might have an important, context-dependent role in breast cancer.


Subject(s)
Breast Neoplasms/classification , Breast Neoplasms/metabolism , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , Mammary Glands, Animal/pathology , Tissue Array Analysis , Animals , Breast Neoplasms/pathology , DNA-Binding Proteins/metabolism , Female , Humans , Hyperplasia , Lactation , Mammary Glands, Animal/metabolism , Mammary Tumor Virus, Mouse/physiology , Mice, Transgenic , Middle Aged , Parity , Precancerous Conditions/pathology , Pregnancy , STAT5 Transcription Factor/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism
10.
J Pathol ; 230(4): 430-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23616343

ABSTRACT

Src family kinase activity is elevated in a number of human cancers including breast cancer. This increased activity has been associated with aggressive disease and poor prognosis. Src inhibitors are currently in clinical development with a number of trials currently assessing their activity in breast cancer. However, the results to date have been disappointing and a further evaluation of the preclinical effects of Src inhibitors is required to help establish whether these agents will be useful in the treatment of breast cancer. In this study we investigate the effects of dasatinib, which is a potent inhibitor of Src family kinases, on the initiation and development of breast cancer in a genetically engineered model of the disease. The mouse model utilized is driven by expression of activated ErbB-2 under the transcriptional control of its endogenous promoter coupled with conditional loss of Pten under the control of Cre recombinase expressed by the BLG promoter. We show that daily oral administration of dasatinib delays tumour onset and increases overall survival but does not inhibit the proliferation of established tumours. The striking difference between the dasatinib-treated group of tumours and the vehicle controls was the prominent squamous metaplasia that was seen in six out of 11 dasatinib-treated tumours. This was accompanied by a dramatic up-regulation of both E-cadherin and ß-catenin and down-regulation of ErbB-2 in the dasatinib-treated tumours. Dasatinib also inhibited both the migration and the invasion of tumour-derived cell lines in vitro. Together these data support the argument that benefits of Src inhibitors may predominate in early or even pre-invasive disease.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , src-Family Kinases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dasatinib , Dose-Response Relationship, Drug , Drug Administration Schedule , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Neoplastic , Genes, erbB-2 , Integrases/genetics , Integrases/metabolism , Lactoglobulins/genetics , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Invasiveness , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction/drug effects , Thiazoles/administration & dosage , Time Factors , beta Catenin/genetics , beta Catenin/metabolism , src-Family Kinases/metabolism
11.
Breast Cancer Res ; 14(6): R143, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23127292

ABSTRACT

INTRODUCTION: The prognostic significance of p53 protein expression in early breast cancer remains uncertain, with some but not all studies finding an association with poorer outcomes. Estrogen receptor (ER) expression is both a positive prognostic marker and predictive of response to endocrine therapies. The relationship between these biomarkers is unknown. METHODS: We constructed tissue microarrays (TMAs) from available pathological material from 1113 patients participating in two randomized clinical trials comparing endocrine therapy alone versus chemo-endocrine therapy in node-negative breast cancer. Expression of p53 defined as >10% positive nuclei was analyzed together with prior immunohistochemical assays of ER performed at central pathological review of whole tumor sections. RESULTS: ER was present (i.e. >1% positive tumor cell nuclei) in 80.1% (880/1092). p53 expression was significantly more frequent when ER was absent, 125/212 (59%) than when ER was present, 171/880 (19%), p <0.0001. A significant qualitative interaction was observed such that p53 expression was associated with better disease-free survival (DFS) and overall survival (OS) among patients whose tumors did not express ER, but worse DFS and OS among patients whose tumors expressed ER. The interaction remained significant after allowance for pathologic variables, and treatment. Similar effects were seen when luminal and non-luminal intrinsic subtypes were compared. CONCLUSIONS: Interpretation of the prognostic significance of p53 expression requires knowledge of concurrent expression of ER. The reason for the interaction between p53 and ER is unknown but may reflect qualitatively different p53 mutations underlying the p53 expression in tumors with or without ER expression. TRIAL REGISTRATION: Current Controlled Trials ACTRN12607000037404 (Trial VIII) and ACTRN12607000029493 (Trial IX).


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Receptors, Estrogen/biosynthesis , Tamoxifen/therapeutic use , Tumor Suppressor Protein p53/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cyclophosphamide/therapeutic use , Disease-Free Survival , Female , Fluorouracil/therapeutic use , Goserelin/therapeutic use , Humans , Lymph Nodes , Methotrexate/therapeutic use , Middle Aged , Tissue Array Analysis , Treatment Outcome
12.
Semin Cell Dev Biol ; 23(5): 567-73, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22426022

ABSTRACT

This review is intended to give an overview of current knowledge on human breast development. It focuses on the limitations of our understanding on the origins of human breast cancer in the context of this mainly morphological and static assessment of what is known about human breast development. The world literature is very limited and caution is needed in drawing analogies with the mouse. There is an increasing emphasis on research to understand normal stem cells in the breast on the assumption that these are the targets for the initiation of breast cancer. It is thus a priority to understand normal human breast development, but there are major obstacles to such studies mainly due to ethical considerations and to tissue acquisition.


Subject(s)
Breast/growth & development , Aging , Animals , Breast/embryology , Breast Neoplasms/metabolism , Humans , Keratins/metabolism , Neoplastic Stem Cells/metabolism
13.
Am J Pathol ; 177(5): 2323-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20847288

ABSTRACT

Reelin is a secreted, signaling protein associated with neuronal cell positioning and migration. Recently, reelin was found to be epigenetically silenced in gastric and pancreatic cancers in which down-regulation was associated with increased migratory ability and reduced survival. Here we analyzed reelin expression by immunohistochemistry in 17 normal breast tissue samples from reduction mammoplasties and in two independent tissue microarrays of 136 and more than 2000 breast cancer biopsy samples, respectively. Results were analyzed with regard to clinical parameters, including BRE (Bloom, Richardson, Elston) grade, nodal status, estrogen receptor and HER2 status, and overall survival. Reelin was expressed in the luminal epithelium and myoepithelium of the normal human breast but not in cancerous breasts. Loss of reelin protein expression correlated significantly with decreased survival (P=0.01) and positive lymph node status (P<0.001). By measuring reelin expression and promoter methylation status in 39 primary breast tumors, as well as in breast cancer-derived cell lines before and after decitabine treatment, we established that reelin expression levels correlated inversely with promoter methylation status, whereas demethylation increased reelin mRNA expression in vitro. Reelin overexpression in MDA-MB231 cells, as well as incubation with recombinant reelin, suppressed cell migration, invadopodia formation, and invasiveness in vitro. We conclude that reelin may play an important role in controlling invasiveness and metastatic potential of breast cancer cells and that its expression is controlled by promoter methylation.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Epigenesis, Genetic , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Line, Tumor , Cell Movement , Collagen Type I/metabolism , Extracellular Matrix Proteins/genetics , Female , HEK293 Cells , Humans , Neoplasm Invasiveness , Nerve Tissue Proteins/genetics , Prognosis , Promoter Regions, Genetic , Reelin Protein , Serine Endopeptidases/genetics
14.
Breast Cancer Res ; 12(4): 310, 2010.
Article in English | MEDLINE | ID: mdl-20828424

ABSTRACT

Cancer vaccines are the Holy Grail for patients and clinicians alike. The possibility that we can be vaccinated against common cancers is very appealing and the socioeconomic consequences are significant. A recent paper from Vincent Tuohy's group, published in the journal Nature Medicine, suggests a new approach for the development of a prophylactic vaccine for breast cancer. Their strategy was to induce mammary gland failure in mice by immunisation with an antibody specific to a milk protein that resulted in autoimmunity during lactation. This also showed some efficacy as a therapeutic vaccine. Can we look forward to the elimination of breast cancer?


Subject(s)
Breast Neoplasms/prevention & control , Cancer Vaccines/therapeutic use , Lactalbumin/immunology , Mammary Neoplasms, Experimental/prevention & control , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Female , Humans , Immunization , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C
15.
J Clin Oncol ; 28(18): 2966-73, 2010 Jun 20.
Article in English | MEDLINE | ID: mdl-20458051

ABSTRACT

PURPOSE: Retrospective studies suggest that primary breast cancers lacking estrogen receptor (ER) and progesterone receptor (PR) and not overexpressing human epidermal growth factor receptor 2 (HER2; triple-negative tumors) are particularly sensitive to DNA-damaging chemotherapy with alkylating agents. PATIENTS AND METHODS: Patients enrolled in International Breast Cancer Study Group Trials VIII and IX with node-negative, operable breast cancer and centrally assessed ER, PR, and HER2 were included (n = 2,257). The trials compared three or six courses of adjuvant classical cyclophosphamide, methotrexate, and fluorouracil (CMF) with or without endocrine therapy versus endocrine therapy alone. We explored patterns of recurrence by treatment according to three immunohistochemically defined tumor subtypes: triple negative, HER2 positive and endocrine receptor absent, and endocrine receptor present. RESULTS: Patients with triple-negative tumors (303 patients; 13%) were significantly more likely to have tumors > 2 cm and grade 3 compared with those in the HER2-positive, endocrine receptor-absent, and endocrine receptor-present subtypes. No clear chemotherapy benefit was observed in endocrine receptor-present disease (hazard ratio [HR], 0.90; 95% CI, 0.74 to 1.11). A statistically significantly greater benefit for chemotherapy versus no chemotherapy was observed in triple-negative breast cancer (HR, 0.46; 95% CI, 0.29 to 0.73; interaction P = .009 v endocrine receptor-present disease). The magnitude of the chemotherapy effect was lower in HER2-positive endocrine receptor-absent disease (HR, 0.58; 95% CI, 0.29 to 1.17; interaction P = .24 v endocrine receptor-present disease). CONCLUSION: The magnitude of benefit of CMF chemotherapy is largest in patients with triple-negative, node-negative breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Lymph Nodes/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Cyclophosphamide/administration & dosage , Female , Fluorouracil/administration & dosage , Humans , Immunoenzyme Techniques , Methotrexate/administration & dosage , Middle Aged , Neoplasm Staging , Retrospective Studies , Survival Rate , Treatment Outcome
16.
Lancet Oncol ; 10(5): 522-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19410197

ABSTRACT

Data suggest that neither our current understanding of the function and signalling of epidermal growth factor receptor (EGFR), nor measurements of receptor expression are reliably predictive of therapeutic responses to EGFR inhibitors. The time has now come to consider whether such poor correlation between receptor expression and clinical response is caused by poor assays or by more fundamental issues relating to the in-vivo function of EGFR. Revisiting some of the early findings of the biology of EGFR function and understanding the limitations of immunohistochemistry as a quantitative technique might provide some clues. However, we still have a lot to learn about this receptor, its many ligands, and its binding partners in normal physiology and disease.


Subject(s)
Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Animals , ErbB Receptors/metabolism , ErbB Receptors/physiology , Humans , Neoplasms/metabolism
17.
J Mammary Gland Biol Neoplasia ; 14(2): 99-116, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19408105

ABSTRACT

Mouse mammary gland involution resembles a wound healing response with suppressed inflammation. Wound healing and inflammation are also associated with tumour development, and a 'wound-healing' gene expression signature can predict metastasis formation and survival. Recent studies have shown that an involuting mammary gland stroma can promote metastasis. It could therefore be hypothesised that gene expression signatures from an involuting mouse mammary gland may provide new insights into the physiological pathways that promote breast cancer progression. Indeed, using the HOPACH clustering method, the human orthologues of genes that were differentially regulated at day 3 of mammary gland involution and showed prolonged expression throughout the first 4 days of involution distinguished breast cancers in the NKI 295 breast cancer dataset with low and high metastatic activity. Most strikingly, genes associated with copper ion homeostasis and with HIF-1 promoter binding sites were the most over-represented, linking this signature to hypoxia. Further, six out of the ten mRNAs with strongest up-regulation in cancers with poor survival code for secreted factors, identifying potential candidates that may be involved in stromal/matrix-enhanced metastasis formation/breast cancer development. This method therefore identified biological processes that occur during mammary gland involution, which may be critical in promoting breast cancer metastasis that could form a basis for future investigation, and supports a role for copper in breast cancer development.


Subject(s)
Breast Neoplasms/genetics , Breast/physiology , Gene Expression Profiling , Lactation/genetics , Mammary Glands, Animal/physiology , Mammary Neoplasms, Experimental/genetics , Neoplasm Metastasis/genetics , RNA, Messenger/analysis , Animals , Breast/metabolism , Breast/pathology , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Ceruloplasmin/genetics , Ceruloplasmin/physiology , Cluster Analysis , Copper/metabolism , Cytoskeletal Proteins/genetics , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , Homeostasis , Humans , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , Stromal Cells/metabolism
18.
Nat Rev Cancer ; 9(2): 128-34, 2009 02.
Article in English | MEDLINE | ID: mdl-19132008

ABSTRACT

It has been proposed that gene expression profiles will revolutionize the classification of breast cancer, eventually replacing histopathology with a more reproducible technology. These new approaches, combined with a better understanding of the cellular origins of breast cancer, should enable us to identify patient subgroups for more effective therapy. However, in such a rapidly advancing field it is essential that initial and thought-provoking results do not become established as 'facts' without question. This Opinion addresses some of the negatives and positives generated by the term 'basal-like' breast cancer, and questions its existence as an entity.


Subject(s)
Breast Neoplasms/genetics , Terminology as Topic , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Breast/anatomy & histology , Breast/physiology , Breast Neoplasms/classification , Breast Neoplasms/metabolism , Female , Gene Expression Profiling , Humans , Keratins/genetics , Keratins/metabolism
19.
J Clin Oncol ; 26(34): 5569-75, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18981464

ABSTRACT

PURPOSE: To evaluate the prognostic and predictive value of Ki-67 labeling index (LI) in a trial comparing letrozole (Let) with tamoxifen (Tam) as adjuvant therapy in postmenopausal women with early breast cancer. PATIENTS AND METHODS: Breast International Group (BIG) trial 1-98 randomly assigned 8,010 patients to four treatment arms comparing Let and Tam with sequences of each agent. Of 4,922 patients randomly assigned to receive 5 years of monotherapy with either agent, 2,685 had primary tumor material available for central pathology assessment of Ki-67 LI by immunohistochemistry and had tumors confirmed to express estrogen receptors after central review. The prognostic and predictive value of centrally measured Ki-67 LI on disease-free survival (DFS) were assessed among these patients using proportional hazards modeling, with Ki-67 LI values dichotomized at the median value of 11%. RESULTS: Higher values of Ki-67 LI were associated with adverse prognostic factors and with worse DFS (hazard ratio [HR; high:low] = 1.8; 95% CI, 1.4 to 2.3). The magnitude of the treatment benefit for Let versus Tam was greater among patients with high tumor Ki-67 LI (HR [Let:Tam] = 0.53; 95% CI, 0.39 to 0.72) than among patients with low tumor Ki-67 LI (HR [Let:Tam] = 0.81; 95% CI, 0.57 to 1.15; interaction P = .09). CONCLUSION: Ki-67 LI is confirmed as a prognostic factor in this study. High Ki-67 LI levels may identify a patient group that particularly benefits from initial Let adjuvant therapy.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents/therapeutic use , Ki-67 Antigen/biosynthesis , Nitriles/therapeutic use , Tamoxifen/therapeutic use , Triazoles/therapeutic use , Breast Neoplasms , Chemotherapy, Adjuvant , Cohort Studies , Endocrine System , Female , Humans , Letrozole , Postmenopause , Predictive Value of Tests , Prognosis , Proportional Hazards Models
20.
J Clin Oncol ; 26(18): 3006-14, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18458044

ABSTRACT

PURPOSE: To determine how patients with infiltrating lobular carcinoma (ILC) differ from patients with the more common infiltrating ductal carcinoma (IDC) with regard to patient and tumor factors, local treatment, and patterns of recurrence. PATIENTS AND METHODS: Twelve thousand two hundred six breast cancer patients entered onto 15 International Breast Cancer Study Group trials between 1978 and 2002 were categorized as having ILC, IDC, or other/mixed types. RESULTS: Seven hundred sixty-seven tumors (6.2%) were classified as ILC, 8,607 (70.5%) were classified as IDC, and 2,832 (23.2%) were classified as other. The analysis is limited to the 9,374 patients categorized as either pure IDC or ILC. The median follow-up time was 13 years. Compared with IDC, ILC was associated with older age; larger, better differentiated, and estrogen receptor (ER)-positive tumors; and less vessel invasion. Mastectomy was used more frequently for ILC (P < .01). There was a significant (P < .01) early advantage in disease-free survival and overall survival for the ILC cohort followed by a significant (P < .01) late advantage for the IDC cohort after 6 and 10 years, respectively. Similar patterns were observed in cohorts defined by ER status. ILC was associated with an increased incidence of bone events but a decrease in regional and lung events (all P < .01). CONCLUSION: ILC is more than a histologic variant of breast cancer. The diagnosis of ILC carries distinct prognostic and biologic implications.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Lobular/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/epidemiology , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/epidemiology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Carcinoma, Lobular/diagnosis , Carcinoma, Lobular/epidemiology , Carcinoma, Lobular/therapy , Clinical Trials as Topic , Disease-Free Survival , Female , Humans , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...