29 results on '"Sinead Cocchiglia"'
Search Results
2. Supplementary Table 1 from Metastatic Progression with Resistance to Aromatase Inhibitors Is Driven by the Steroid Receptor Coactivator SRC-1
- Author
-
Leonie S. Young, Arnold D.K. Hill, Paul Tibbitts, Jane O'Hara, Stephen Sande, Sinead Cocchiglia, Eamon Hughes, Christopher Byrne, Sarah M. Theissen, and Jean McBryan
- Abstract
PDF file - 28K
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 1 from Metastatic Progression with Resistance to Aromatase Inhibitors Is Driven by the Steroid Receptor Coactivator SRC-1
- Author
-
Leonie S. Young, Arnold D.K. Hill, Paul Tibbitts, Jane O'Hara, Stephen Sande, Sinead Cocchiglia, Eamon Hughes, Christopher Byrne, Sarah M. Theissen, and Jean McBryan
- Abstract
PDF file - 175K
- Published
- 2023
- Full Text
- View/download PDF
4. O28: M6A DEMETHYLASE FTO A POTENTIAL TARGET IN BRAIN METASTATIC BREAST CANCER
- Author
-
Stephen Keelan, Fiona T Bane, Leonie S. Young, Sara Charmsaz, Damir Varešlija, Sinead Cocchiglia, Siobhan Purcell, and A Hill
- Subjects
biology ,business.industry ,Disease progression ,medicine.disease ,FTO gene ,Metastatic breast cancer ,Breast cancer ,Tumor progression ,Cancer research ,medicine ,biology.protein ,Demethylase ,Surgery ,Epigenetics ,Biological response modifiers ,business - Abstract
Introduction Brain metastasis (BrM) occurs in 10-30% of patients with advanced breast cancer (BC). BrM is increasing in incidence and confers a poor prognosis. We aimed to investigate the contribution of global epi-transcriptomic alterations in N6-methyladenosine (m6A) RNA-methylation as a therapeutic target in brain metastatic breast cancer. Method In preliminary studies we have demonstrated m6A demethylase – FTO as the main contributor to the progression of ER+ breast cancer. Furthermore an association between FTO and reduced disease-free-survival (n=870, p=0.018) was observed. Here we conducted an epigenetic inhibitor screen using two therapeutic agents, ethyl-ester-meclofenamic acid (MA2) and FB23-2 on matched 2D cell line, 3D organoid cultures and patient-derived xenografts (PDX) explant models of brain metastasis. Result Upon integration of mapped global RNA methylation landscape with matched proteomic analysis, we observed genome-wide RNA hypo-methylation of key pluripotency genes, including SOX2 and KLF4, as key players underlying tumour progression to the brain. Genetic and pharmacological inhibition of FTO in novel ex vivo models of BrM significantly reduced protein expression levels of KLF4 and SOX2. Moreover, pharmacological inhibition of FTO with MA2 and FB23-2, inhibited cell proliferation in endocrine-resistant BC and patient BrM cells. We translate our findings to the clinic by demonstrating the efficacy of anti-FTO therapies in several unique PDX and 3D organoid BrM models. Conclusion Our results reveal epi-transcriptional remodelling events as a key mechanism in BrM. This study establishes an early role for targeting RNA methylation in the management of disease progression and presents FTO as a potential therapeutic target in BrM. Take-home message This study establishes an early role for targeting RNA methylation in the management of disease progression and presents FTO as a potential therapeutic target in brain metastatic breast cancer.
- Published
- 2021
- Full Text
- View/download PDF
5. O12: IHC ANALYSIS OF RESIDUAL DISEASE POST NEOADJUVANT TRASTUZUMAB FOR EARLY STAGE BREAST CANCER REVEALS ER/HER2 CROSSTALK THROUGH INCREASED ER SIGNALLING
- Author
-
Nicola Cosgrove, Michael Flanagan, Katherine M. Sheehan, Sinead Cocchiglia, Leonie S. Young, Siobhan Purcell, Fiona T Bane, D Varšlija, A Hill, and Sara Charmsaz
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Disease ,medicine.disease ,Crosstalk (biology) ,Breast cancer ,Signalling ,Trastuzumab ,Internal medicine ,medicine ,Immunohistochemistry ,Surgery ,Stage (cooking) ,skin and connective tissue diseases ,business ,medicine.drug - Abstract
Introduction Therapeutic pressure functionally affects oncogenes and related signalling pathways through dynamic alterations in transcriptional and epigenetic alterations. Altered receptor status occurs throughout tumour progression and may be influenced by adjuvant and neoadjuvant therapies. Recurrent transcriptional remodelling events have been described in the progression of primary breast cancer to metastasis, including increased tyrosine kinase signalling, specifically Her2, and loss of ESR1 gene expression. We hypothesise that in the setting of tyrosine kinase inhibition, an increase in estrogen receptor (ER) signalling is observed. Method A database of patients recruited to ICORG trial 07/09 was queried to identify patients with histologically confirmed, Her2-overexpressing or Her2 amplified, nonmetastatic, invasive breast cancer who received neoadjuvant trastuzumab, alone or in combination with neoadjuvant systemic chemotherapy. Clinicopathological characteristics recorded include age at diagnosis, clinical stage, receptor status and percentage positivity, and pathological complete response. Result A total of 55 patients identified on ICORG trial 09/07 received neoadjuvant trastuzumab. Of these, 27 achieved a complete pathological response (49%; n=27/55). In those with residual disease, a gain in mean ER staining percentage positivity was observed in the residual disease compared to diagnostic biopsy staining (59.22 vs 45.11; p=0.03). A corresponding loss in Her2 percentage staining positivity was also observed (p=0.006). Conclusion An inverse correlation was observed between loss of Her2 positivity and percentage gain in ER staining in patients with residual disease following treatment with neoadjuvant trastuzumab. Further study is needed to elucidate the regulatory mechanism of ER/Her2 crosstalk, which may be epigenetically regulated through DNA methylation. Take-home message ER/Her2 crosstalk can be demonstrated clinically in IHC analysis of patients with residual disease post neoadjuvant trastuzumab. Tyrosine kinase inhibition in the form of neoadjuvant trastuzumab results in loss of Her2 signalling and corresponding gain in ER signalling.
- Published
- 2021
- Full Text
- View/download PDF
6. O50: DEVELOPMENT OF A PATIENT-DERIVED TUMOUR ORGANOIDS FROM METASTATIC BREAST CANCER FOR ASSESSMENT OF NOVEL CLINICALLY ACTIONABLE TARGETS
- Author
-
Francesca Brett, Lance Hudson, P Jagust, Sinead Cocchiglia, Leonie S. Young, Damir Varešlija, Jane Cryan, A Hill, MO Dablouk, PJ O'Haloran, and Alan Beausang
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Organoid ,Medicine ,Surgery ,business ,medicine.disease ,Metastatic breast cancer - Abstract
Introduction Metastatic breast cancer (MBC) is the main source of mortality in breast cancer patients largely due to lack of effective treatments. Our previous results suggest that tumour transcriptional heterogeneity drives therapy resistance and cancer relapse. While traditional in vitro human cancer cell line models have been widely used for disease modelling, they do not faithfully recapitulate the pathophysiology of MBC. Method In this study we developed patient-derived tumour organoid cultures from frozen patient-derived (PDX) models of MBC. Using those models we performed preclinical drug screening of investigational and FDA approved therapeutics previously uncovered by us as potentially clinically actionable in MBC. Result Our results reveal high heterogeneity in the responses to various targeted therapies among tested MBC organoids, which makes them a valuable tool for studying intra-tumor heterogeneity and drug response. Moreover, drug screening identified a divergent set of the breast to brain metastatic MBC organoids that showed high sensitivity to a new class of tyrosine kinase receptors, RET. Conclusion Taken together, our novel MBC models and methodology applied here provides an important modelling tool to assess the contribution of intra-tumour heterogeneity and microenvironment to drug response as they recapitulate the cellular, structural and biochemical complexity previously observed in our genomic characterisation of MBCs. Application of this type of translational research will enhance the development of new targeted precision medicine strategies and prelude stratification for clinical trials. Abbreviations MBC- Metastatic Breast Cancer; PDX- Patient-Derived Xenografts; FDA- Food and Drug Administration; RET- Receptor Tyrosine Kinase Take-home message Patient-derived tumour organoid cultures provide an important modelling tool to assess the contribution of intra-tumour heterogeneity and microenvironment to drug response as they recapitulate the cellular, structural and biochemical complexity previously observed in genomic characterisation of metastatic breast cancer. SURGICAL EDUCATION AND TRAINING
- Published
- 2021
- Full Text
- View/download PDF
7. Comparative analysis of the AIB1 interactome in breast cancer reveals MTA2 as a repressive partner which silences E-Cadherin to promote EMT and associates with a pro-metastatic phenotype
- Author
-
Jane Cryan, Siobhan Purcell, Andrew H. Sims, Philip J. O’Halloran, Francesca Brett, Adrian V. Lee, Arran K Turnbul, Fiona T Bane, Lance Hudson, Jason S. Carroll, Elspeth Ward, Nolan Priedigkeit, Leonie S. Young, J Michael Dixon, Steffi Oesterreich, Nicola Cosgrove, Damir Varešlija, Sinead Cocchiglia, Michael Farrell, Arnold D.K. Hill, Aisling M. Redmond, Alan Beausang, Sara Charmsaz, Varešlija, Damir [0000-0003-1000-0357], Ward, Elspeth [0000-0002-7530-2279], Cocchiglia, Sinéad [0000-0002-1596-1950], Oesterreich, Steffi [0000-0002-2537-6923], Lee, Adrian V [0000-0001-9917-514X], Sims, Andrew H [0000-0001-9082-3665], Carroll, Jason S [0000-0003-3643-0080], Young, Leonie S [0000-0002-4904-0367], Apollo - University of Cambridge Repository, Lee, Adrian V. [0000-0001-9917-514X], Sims, Andrew H. [0000-0001-9082-3665], Carroll, Jason S. [0000-0003-3643-0080], and Young, Leonie S. [0000-0002-4904-0367]
- Subjects
0301 basic medicine ,Cancer Research ,Estrogen receptor ,CDH1 ,Metastasis ,Nuclear Receptor Coactivator 3 ,0302 clinical medicine ,Breast cancer ,Neoplasm Metastasis ,skin and connective tissue diseases ,13/89 ,biology ,Cadherins ,Prognosis ,Gene Expression Regulation, Neoplastic ,631/67/322 ,Phenotype ,030220 oncology & carcinogenesis ,MCF-7 Cells ,38/39 ,64/60 ,Female ,Epithelial-Mesenchymal Transition ,Breast Neoplasms ,Article ,Disease-Free Survival ,Histone Deacetylases ,38/91 ,03 medical and health sciences ,Antigens, CD ,Coactivator ,Genetics ,medicine ,Humans ,Epithelial–mesenchymal transition ,Molecular Biology ,Cell Proliferation ,96/106 ,82/58 ,Estrogen Receptor alpha ,631/67/1347 ,medicine.disease ,Repressor Proteins ,Tamoxifen ,030104 developmental biology ,Nuclear receptor ,Nuclear receptor coactivator 3 ,Cancer cell ,13/51 ,biology.protein ,Cancer research - Abstract
Funder: Breast Cancer Ireland GR 14-0883, Steroid regulated cancer cells use nuclear receptors and associated regulatory proteins to orchestrate transcriptional networks to drive disease progression. In primary breast cancer, the coactivator AIB1 promotes estrogen receptor (ER) transcriptional activity to enhance cell proliferation. The function of the coactivator in ER+ metastasis however is not established. Here we describe AIB1 as a survival factor, regulator of pro-metastatic transcriptional pathways and a promising actionable target. Genomic alterations and functional expression of AIB1 associated with reduced disease-free survival in patients and enhanced metastatic capacity in novel CDX and PDX ex-vivo models of ER+ metastatic disease. Comparative analysis of the AIB1 interactome with complementary RNAseq characterized AIB1 as a transcriptional repressor. Specifically, we report that AIB1 interacts with MTA2 to form a repressive complex, inhibiting CDH1 (encoding E-cadherin) to promote EMT and drive progression. We further report that pharmacological and genetic inhibition of AIB1 demonstrates significant anti-proliferative activity in patient-derived models establishing AIB1 as a viable strategy to target endocrine resistant metastasis. This work defines a novel role for AIB1 in the regulation of EMT through transcriptional repression in advanced cancer cells with a considerable implication for prognosis and therapeutic interventions.
- Published
- 2021
8. AB069. SOH22ABS209. The epitranscriptomic landscape in estrogen receptor (ER)-positive breast cancer disease progression
- Author
-
Stephen Keelan, Mihaela Ola, Sara Charmsaz, Sinead Cocchiglia, Karen Crowley, Siobhan Purcell, Fiona Bane, Aisling Hegarty, Ben Doherty, Katherine Sheehan, Lance Hudson, Nicola Cosgrove, Benjamin Roux, Muriel Laine, Geoffrey Greene, Damir Vareslija, Arnold Hill, and Leonie Young
- Subjects
General Medicine - Published
- 2022
- Full Text
- View/download PDF
9. S100β as a serum marker in endocrine resistant breast cancer
- Author
-
Roisin M. Dwyer, Paul Tibbitts, Christopher Byrne, Leonie S. Young, Arnold D.K. Hill, Sinead Cocchiglia, Sara Charmsaz, Bryan T. Hennessy, E Hughes, Fiona T Bane, Jean McBryan, Marie McIlroy, and Michael J. Kerin
- Subjects
0301 basic medicine ,Oncology ,Estrogen receptor ,lcsh:Medicine ,Mice ,0302 clinical medicine ,Breast cancer ,dasatinib ,S100β ,estrogen-receptor ,Aged, 80 and over ,Standard treatment ,General Medicine ,Middle Aged ,Dasatinib ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Biomarker (medicine) ,Immunohistochemistry ,Female ,transcription ,Signal Transduction ,Research Article ,medicine.drug ,Endocrine resistance ,Adult ,medicine.medical_specialty ,Antineoplastic Agents, Hormonal ,kinase inhibitor ,s100 beta ,predictor ,Breast Neoplasms ,Enzyme-Linked Immunosorbent Assay ,S100 Calcium Binding Protein beta Subunit ,Disease-Free Survival ,03 medical and health sciences ,Cell Line, Tumor ,Internal medicine ,melanoma ,medicine ,Animals ,Humans ,Endocrine system ,Aged ,therapy ,business.industry ,lcsh:R ,src-1 ,Biomarker ,medicine.disease ,Xenograft Model Antitumor Assays ,Tamoxifen ,030104 developmental biology ,Endocrinology ,Drug Resistance, Neoplasm ,Tissue Array Analysis ,Neoplasm Recurrence, Local ,steroid-receptor coactivator-1 ,business ,Biomarkers - Abstract
Background Endocrine therapy is standard treatment for estrogen receptor (ER)-positive breast cancer. However, its efficacy is limited by intrinsic and acquired resistance. Here the potential of S100β as a biomarker and inhibition of its signaling network as a therapeutic strategy in endocrine treated patients was investigated. Methods The expression of S100β in tissue and serum was assessed by immunohistochemistry and an enzyme-linked immunosorbent assay, respectively. The S100β signaling network was investigated in cell line models of endocrine resistance by western blot, PCR, immunoprecipitation, and chromatin-immunoprecipitation. Endocrine resistant xenografts and tumor explants from patients with resistant tumors were treated with endocrine therapy in the presence and absence of the p-Src kinase inhibitor, dasatinib. Results Tissue and serum levels of S100β were found to predict poor disease-free survival in endocrine-treated patients (n = 509, HR 2.32, 95% CI is 1.58–3.40, p
- Published
- 2017
- Full Text
- View/download PDF
10. ADAM22/LGI1 complex as a new actionable target for breast cancer brain metastasis
- Author
-
Matteo Battaglini, Chiara Martinelli, Adrian V. Lee, Arnold D.K. Hill, Fiona T Bane, Stephen F. Madden, Siobhan Purcell, Katherine M. Sheehan, Ricardo Marques, Francesca Brett, Attilio Marino, Gianni Ciofani, Stephen Keelan, Kieran Brennan, Sara Charmsaz, Christopher Byrne, Jarlath C. Bolger, Sinead Cocchiglia, Ben Doherty, Ann M. Hopkins, Petra Jagust, Damir Varešlija, Nicola Cosgrove, Nolan Priedigkeit, Leonie S. Young, Philip J. O’Halloran, and Steffi Oesterreich
- Subjects
0301 basic medicine ,medicine.medical_treatment ,ADAM22 ,lcsh:Medicine ,Breast Neoplasms ,Nerve Tissue Proteins ,Metastasis ,Targeted therapy ,Blood–brain barrier ,03 medical and health sciences ,ECM signalling ,0302 clinical medicine ,Breast cancer ,In vivo ,Biomimetic Materials ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,business.industry ,Brain Neoplasms ,Gene Expression Profiling ,lcsh:R ,Breast cancer metastases ,Intracellular Signaling Peptides and Proteins ,Cancer ,Brain metastases ,General Medicine ,medicine.disease ,Metastatic breast cancer ,3. Good health ,ADAM Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,LGI1 ,Neoplasm Recurrence, Local ,business ,Peptides ,Ex vivo ,Brain metastasis ,Research Article - Abstract
Background Metastatic breast cancer is a major cause of cancer-related deaths in woman. Brain metastasis is a common and devastating site of relapse for several breast cancer molecular subtypes, including oestrogen receptor-positive disease, with life expectancy of less than a year. While efforts have been devoted to developing therapeutics for extra-cranial metastasis, drug penetration of blood–brain barrier (BBB) remains a major clinical challenge. Defining molecular alterations in breast cancer brain metastasis enables the identification of novel actionable targets. Methods Global transcriptomic analysis of matched primary and metastatic patient tumours (n = 35 patients, 70 tumour samples) identified a putative new actionable target for advanced breast cancer which was further validated in vivo and in breast cancer patient tumour tissue (n = 843 patients). A peptide mimetic of the target’s natural ligand was designed in silico and its efficacy assessed in in vitro, ex vivo and in vivo models of breast cancer metastasis. Results Bioinformatic analysis of over-represented pathways in metastatic breast cancer identified ADAM22 as a top ranked member of the ECM-related druggable genome specific to brain metastases. ADAM22 was validated as an actionable target in in vitro, ex vivo and in patient tumour tissue (n = 843 patients). A peptide mimetic of the ADAM22 ligand LGI1, LGI1MIM, was designed in silico. The efficacy of LGI1MIM and its ability to penetrate the BBB were assessed in vitro, ex vivo and in brain metastasis BBB 3D biometric biohybrid models, respectively. Treatment with LGI1MIM in vivo inhibited disease progression, in particular the development of brain metastasis. Conclusion ADAM22 expression in advanced breast cancer supports development of breast cancer brain metastasis. Targeting ADAM22 with a peptide mimetic LGI1MIM represents a new therapeutic option to treat metastatic brain disease.
- Published
- 2020
11. Network analysis of SRC-1 reveals a novel transcription factor hub which regulates endocrine resistant breast cancer
- Author
-
Lance Hudson, Nicola Cosgrove, Alacoque Browne, Arnold D.K. Hill, Fiona T Bane, Elspeth Ward, Ailis Fagan, Sinead Cocchiglia, Siobhan Purcell, Damir Varešlija, Jason S. Carroll, Sara Charmsaz, Leonie S. Young, Aisling M. Redmond, Varešlija, Damir [0000-0003-1000-0357], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Transcriptional Activation ,Cancer Research ,Antineoplastic Agents, Hormonal ,Gene regulatory network ,Estrogen receptor ,Breast Neoplasms ,Mice, SCID ,Biology ,Article ,03 medical and health sciences ,Mice ,Nuclear Receptor Coactivator 1 ,Transcription (biology) ,Mice, Inbred NOD ,Genetics ,Tumor Cells, Cultured ,Animals ,Humans ,Gene Regulatory Networks ,Molecular Biology ,Transcription factor ,Regulation of gene expression ,Gene Expression Profiling ,Chromatin Assembly and Disassembly ,Microarray Analysis ,Cell biology ,Nuclear receptor coactivator 1 ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Nuclear receptor ,NFIA ,Drug Resistance, Neoplasm ,MCF-7 Cells ,Female ,Transcriptome - Abstract
Steroid receptor coactivator 1 (SRC-1) interacts with nuclear receptors and other transcription factors (TFs) to initiate transcriptional networks and regulate downstream genes which enable the cancer cell to evade therapy and metastasise. Here we took a top–down discovery approach to map out the SRC-1 transcriptional network in endocrine resistant breast cancer. First, rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) was employed to uncover new SRC-1 TF partners. Next, RNA sequencing (RNAseq) was undertaken to investigate SRC-1 TF target genes. Molecular and patient-derived xenograft studies confirmed STAT1 as a new SRC-1 TF partner, important in the regulation of a cadre of four SRC-1 transcription targets, NFIA, SMAD2, E2F7 and ASCL1. Extended network analysis identified a downstream 79 gene network, the clinical relevance of which was investigated in RNAseq studies from matched primary and local-recurrence tumours from endocrine resistant patients. We propose that SRC-1 can partner with STAT1 independently of the estrogen receptor to initiate a transcriptional cascade and control regulation of key endocrine resistant genes.
- Published
- 2018
12. Epigenome-wide SRC-1 mediated gene silencing represses cellular differentiation in advanced breast cancer
- Author
-
Arnold D.K. Hill, Siobhan Purcell, Nicola Cosgrove, Alacoque Browne, Elspeth Ward, Sudipto Das, Damir Varešlija, Andrew H. Sims, Leonie S. Young, Sinead Cocchiglia, Phillip J O'Halloran, Ailis Fagan, Sara Charmsaz, Lance Hudson, and Darran P. O'Connor
- Subjects
0301 basic medicine ,Epigenomics ,Cancer Research ,Cellular differentiation ,Estrogen receptor ,Breast Neoplasms ,Biology ,Disease-Free Survival ,Metastasis ,03 medical and health sciences ,Breast cancer ,breast cancer ,medicine ,Humans ,metastasis ,Neoplasm Invasiveness ,SRC-1 ,Epigenetics ,Breast ,Gene Silencing ,Neoplasm Metastasis ,Cell Proliferation ,DNA methylation ,Cancer ,endocrine resistant breast cancer ,Cell Differentiation ,Epigenome ,DNA Methylation ,medicine.disease ,Microarray Analysis ,hormone-dependent cancer ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,src-Family Kinases ,Oncology ,Receptors, Estrogen ,Drug Resistance, Neoplasm ,Cancer research ,MCF-7 Cells ,Heterografts ,Female ,CRISPR-Cas Systems ,estrogen receptor - Abstract
Purpose: Despite the clinical utility of endocrine therapies for estrogen receptor–positive (ER) breast cancer, up to 40% of patients eventually develop resistance, leading to disease progression. The molecular determinants that drive this adaptation to treatment remain poorly understood. Methylome aberrations drive cancer growth yet the functional role and mechanism of these epimutations in drug resistance are poorly elucidated. Experimental Design: Genome-wide multi-omics sequencing approach identified a differentially methylated hub of prodifferentiation genes in endocrine resistant breast cancer patients and cell models. Clinical relevance of the functionally validated methyl-targets was assessed in a cohort of endocrine-treated human breast cancers and patient-derived ex vivo metastatic tumors. Results: Enhanced global hypermethylation was observed in endocrine treatment resistant cells and patient metastasis relative to sensitive parent cells and matched primary breast tumor, respectively. Using paired methylation and transcriptional profiles, we found that SRC-1–dependent alterations in endocrine resistance lead to aberrant hypermethylation that resulted in reduced expression of a set of differentiation genes. Analysis of ER-positive endocrine-treated human breast tumors (n = 669) demonstrated that low expression of this prodifferentiation gene set significantly associated with poor clinical outcome (P = 0.00009). We demonstrate that the reactivation of these genes in vitro and ex vivo reverses the aggressive phenotype. Conclusions: Our work demonstrates that SRC-1-dependent epigenetic remodeling is a ’high level’ regulator of the poorly differentiated state in ER-positive breast cancer. Collectively these data revealed an epigenetic reprograming pathway, whereby concerted differential DNA methylation is potentiated by SRC-1 in the endocrine resistant setting. Clin Cancer Res; 24(15); 3692–703. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
13. Transcriptomic Profiling of Sequential Tumors from Breast Cancer Patients Provides a Global View of Metastatic Expression Changes Following Endocrine Therapy
- Author
-
Damir Varešlija, Fiona T Bane, Marie McIlroy, Paul Tibbitts, Peadar Ó Gaora, Lance Hudson, Christopher Byrne, Leonie S. Young, Sinead Cocchiglia, Ailis Fagan, Arnold D.K. Hill, Jean McBryan, Damian McCartan, and Jarlath C. Bolger
- Subjects
Adult ,Cancer Research ,Pathology ,medicine.medical_specialty ,Antineoplastic Agents, Hormonal ,Breast Neoplasms ,Cell Communication ,Disease ,Biology ,Transcriptome ,Mice ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Cluster Analysis ,Humans ,Neoplasm ,Endocrine system ,Gene Regulatory Networks ,Neoplasm Metastasis ,Gene Expression Profiling ,Liver Neoplasms ,Computational Biology ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,Disease Models, Animal ,Treatment Outcome ,Oncology ,Drug Resistance, Neoplasm ,Cancer research ,Female ,Neoplasm Grading ,Biomarkers ,Hormone - Abstract
Purpose: Disease recurrence is a common problem in breast cancer and yet the mechanisms enabling tumor cells to evade therapy and colonize distant organs remain unclear. We sought to characterize global expression changes occurring with metastatic disease progression in the endocrine-resistant setting. Experimental Design: Here, for the first time, RNAsequencing has been performed on matched primary, nodal, and liver metastatic tumors from tamoxifen-treated patients following disease progression. Expression of genes commonly elevated in the metastases of sequenced patients was subsequently examined in an extended matched patient cohort with metastatic disease from multiple sites. The impact of tamoxifen treatment on endocrine-resistant tumors in vivo was investigated in a xenograft model. Results: The extent of patient heterogeneity at the gene level was striking. Less than 3% of the genes differentially expressed between sequential tumors were common to all patients. Larger divergence was observed between primary and liver tumors than between primary and nodal tumors, reflecting both the latency to disease progression and the genetic impact of intervening therapy. Furthermore, an endocrine-resistant in vivo mouse model demonstrated that tamoxifen treatment has the potential to drive disease progression and establish distant metastatic disease. Common functional pathways altered during metastatic, endocrine-resistant progression included extracellular matrix receptor interactions and focal adhesions. Conclusions: This novel global analysis highlights the influence of primary tumor biology in determining the transcriptomic profile of metastatic tumors, as well as the need for adaptations in cell–cell communications to facilitate successful tumor cell colonization of distant host organs. Clin Cancer Res; 21(23); 5371–9. ©2015 AACR.
- Published
- 2015
- Full Text
- View/download PDF
14. Transcriptome Characterization of Matched Primary Breast and Brain Metastatic Tumors to Detect Novel Actionable Targets
- Author
-
Ryan J. Hartmaier, Darran P. O'Connor, Siobhan Purcell, Nicola Cosgrove, Michael Farrell, Philip J. O’Halloran, Adam Brufsky, Li Zhu, Roisin M. Dwyer, Sinead Cocchiglia, Elspeth Ward, Ailis Fagan, Jose Pablo Leone, Aju Mathew, Leonie S. Young, Arnold D.K. Hill, Ronald L. Hamilton, Adrian V. Lee, Nolan Priedigkeit, Steffi Oesterreich, Lance Hudson, Sudipto Das, Damir Varešlija, George C. Tseng, Shannon Puhalla, Peter C. Lucas, Carlos A. Castro, Ahmed Basudan, and Patrick Buckley
- Subjects
Adult ,Cancer Research ,Breast Neoplasms ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Breast cancer ,Gene expression ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Longitudinal Studies ,Biological response modifiers ,Regulation of gene expression ,business.industry ,Brain Neoplasms ,Gene Expression Profiling ,Articles ,Middle Aged ,medicine.disease ,Prognosis ,Xenograft Model Antitumor Assays ,Fold change ,3. Good health ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Oncology ,030220 oncology & carcinogenesis ,Case-Control Studies ,Cancer research ,Female ,Neoplasm Recurrence, Local ,Erratum ,business ,AcademicSubjects/MED00010 ,Ex vivo ,Follow-Up Studies - Abstract
BackgroundBreast cancer brain metastases (BrMs) are defined by complex adaptations to both adjuvant treatment regimens and the brain microenvironment. Consequences of these alterations remain poorly understood, as does their potential for clinical targeting. We utilized genome-wide molecular profiling to identify therapeutic targets acquired in metastatic disease.MethodsGene expression profiling of 21 patient-matched primary breast tumors and their associated brain metastases was performed by TrueSeq RNA-sequencing to determine clinically actionable BrM target genes. Identified targets were functionally validated using small molecule inhibitors in a cohort of resected BrM ex vivo explants (n = 4) and in a patient-derived xenograft (PDX) model of BrM. All statistical tests were two-sided.ResultsConsiderable shifts in breast cancer cell-specific gene expression profiles were observed (1314 genes upregulated in BrM; 1702 genes downregulated in BrM; DESeq; fold change > 1.5, Padj < .05). Subsequent bioinformatic analysis for readily druggable targets revealed recurrent gains in RET expression and human epidermal growth factor receptor 2 (HER2) signaling. Small molecule inhibition of RET and HER2 in ex vivo patient BrM models (n = 4) resulted in statistically significantly reduced proliferation (P < .001 in four of four models). Furthermore, RET and HER2 inhibition in a PDX model of BrM led to a statistically significant antitumor response vs control (n = 4, % tumor growth inhibition [mean difference; SD], anti-RET = 86.3% [1176; 258.3], P < .001; anti-HER2 = 91.2% [1114; 257.9], P < .01).ConclusionsRNA-seq profiling of longitudinally collected specimens uncovered recurrent gene expression acquisitions in metastatic tumors, distinct from matched primary tumors. Critically, we identify aberrations in key oncogenic pathways and provide functional evidence for their suitability as therapeutic targets. Altogether, this study establishes recurrent, acquired vulnerabilities in BrM that warrant immediate clinical investigation and suggests paired specimen expression profiling as a compelling and underutilized strategy to identify targetable dependencies in advanced cancers.
- Published
- 2017
15. Dynamic epi‐transcriptomic landscape mapping with disease progression in estrogen receptor‐positive breast cancer
- Author
-
Stephen Keelan, Mihaela Ola, Sara Charmsaz, Sinéad Cocchiglia, Daniela Ottaviani, Seán Hickey, Siobhan Purcell, Fiona Bane, Aisling Hegarty, Ben Doherty, Katherine Sheehan, Lance Hudson, Nicola Cosgrove, Benjamin Roux, Muriel Laine, Geoffrey Greene, Damir Varešlija, Arnold Konrad Hill, and Leonie Young
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
16. Global Gene Repression by the Steroid Receptor Coactivator SRC-1 Promotes Oncogenesis
- Author
-
Ailis Fagan, Marie McIlroy, Arnold D.K. Hill, Li Qin, Damian McCartan, Leonie S. Young, Sinead Cocchiglia, Peadar O'Gaora, Jarlath C. Bolger, Jianming Xu, Christopher Byrne, Claire A. Walsh, Yuan Hao, and Aoife Cahalin
- Subjects
Jumonji Domain-Containing Histone Demethylases ,Cancer Research ,Carcinogenesis ,PAWR ,Down-Regulation ,Estrogen receptor ,Breast Neoplasms ,Kaplan-Meier Estimate ,Biology ,medicine.disease_cause ,Disease-Free Survival ,Mice ,Nuclear Receptor Coactivator 1 ,Coactivator ,medicine ,Transcriptional regulation ,Animals ,Humans ,Gene Silencing ,Promoter Regions, Genetic ,skin and connective tissue diseases ,Homeodomain Proteins ,Mice, Knockout ,Regulation of gene expression ,Binding Sites ,CD24 Antigen ,Gene Expression Regulation, Neoplastic ,Oncology ,Nuclear receptor coactivator 3 ,MCF-7 Cells ,Cancer research ,Female ,Apoptosis Regulatory Proteins ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Transcriptional control is the major determinant of cell fate. The steroid receptor coactivator (SRC)-1 enhances the activity of the estrogen receptor in breast cancer cells, where it confers cell survival benefits. Here, we report that a global analysis of SRC-1 target genes suggested that SRC-1 also mediates transcriptional repression in breast cancer cells. Combined SRC-1 and HOXC11 ChIPseq analysis identified the differentiation marker, CD24, and the apoptotic protein, PAWR, as direct SRC-1/HOXC11 suppression targets. Reduced expression of both CD24 and PAWR was associated with disease progression in patients with breast cancer, and their expression was suppressed in metastatic tissues. Investigations in endocrine-resistant breast cancer cell lines and SRC-1−/−/PyMT mice confirmed a role for SRC-1 and HOXC11 in downregulation of CD24 and PAWR. Through bioinformatic analysis and liquid chromatography/mass spectrometry, we identified AP1 proteins and Jumonji domain containing 2C (JMD2C/KDM4C), respectively, as members of the SRC-1 interactome responsible for transcriptional repression. Our findings deepen the understanding of how SRC-1 controls transcription in breast cancers. Cancer Res; 74(9); 2533–44. ©2014 AACR.
- Published
- 2014
- Full Text
- View/download PDF
17. PO-507 ADAM22 as a therapeutic target for endocrine resistant metastatic breast cancer
- Author
-
Damir Varešlija, Arnold D.K. Hill, Sinead Cocchiglia, Ben Doherty, Sara Charmsaz, Jarlath C. Bolger, and Leonie S. Young
- Subjects
Cancer Research ,business.industry ,Cancer ,medicine.disease ,Metastatic breast cancer ,Metastasis ,Breast cancer ,Oncology ,Nuclear receptor ,Coactivator ,medicine ,Cancer research ,Biomarker (medicine) ,Endocrine system ,business - Abstract
Introduction The development of endocrine resistance is a major hurdle for the treatment of oestrogen receptor positive (ER+) breast cancer. One of the key contributors to this resistant phenotype is overexpression of the nuclear receptor coactivator SRC-1, which in turn has been shown to promote metastatic disease. Using CHIP-seq and microarray studies, the neuronal protein ADAM22 (A Disintegrin And Metalloproteinase) was identified as a potential pro-metastatic SRC-1 regulated gene in endocrine resistant cell lines. ADAM22 expression promotes both migration and de-differentiation, key hallmarks of metastasis. While clinically, elevated ADAM22 expression predicts poor disease free survival. Here, both the role of ADAM22 in metastatic development and the clinical potential of ADAM22 as a biomarker and therapeutic target were explored. Material and methods The functional effect of ADAM22 modulation on anchorage independent growth and mammosphere formation was investigated using CRISPR/Cas9 knockout and lentiviral overexpression in the endocrine resistant LY2 cell line. To examine the clinical relevance of ADAM22 as a biomarker, ADAM22 expression was examined in matched primary breast and metastatic cancer tissues. A small peptide mimetic of the ADAM22 endogenous ligand LGI1 (LGIMIM) was designed and explored as a potential anti-ADAM22 therapeutic both in vitro and in vivo . Results and discussions ADAM22 expression promotes anchorage independent growth and mammosphere formation, suggesting ADAM22 contributes to both the survival of metastatic cells and their ability to colonise distant sites. ADAM22 expression was significantly higher in primary breast tumours which went on to metastasise versus primaries which did not. A significant increase in ADAM22 expression was found in brain metastatic tissue compared to matched primary tissue, suggesting ADAM22 may prime endocrine resistant cancer cells to colonise the brain. LGIMIM treatment was sufficient to inhibit ADAM22 mediated phenotypes in vitro and to reduce metastatic burden in a xenograft model of endocrine resistance in vivo . Conclusion ADAM22 is a promising biomarker and therapeutic target for endocrine resistant breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
18. Growth factor receptor/steroid receptor cross talk in trastuzumab-treated breast cancer
- Author
-
D. Collins, Paul Tibbitts, Sinead Cocchiglia, A Hill, Leonie S. Young, G Solon, Alex J Eustace, Achim Treumann, Bryan T. Hennessy, Fiona T Bane, and Jean McBryan
- Subjects
Cancer Research ,medicine.medical_specialty ,Cell signaling ,Receptor, ErbB-2 ,Antineoplastic Agents ,Breast Neoplasms ,Biology ,Antibodies, Monoclonal, Humanized ,Proto-Oncogene Proteins c-myc ,Breast cancer ,Growth factor receptor ,Trastuzumab ,Internal medicine ,Survivin ,Genetics ,medicine ,Humans ,Nuclear Receptor Co-Repressor 2 ,skin and connective tissue diseases ,Protein Kinase Inhibitors ,neoplasms ,Molecular Biology ,Transcription factor ,Tumor Suppressor Proteins ,Receptor Cross-Talk ,medicine.disease ,Endocrinology ,Receptors, Estrogen ,Monoclonal ,MCF-7 Cells ,Cancer research ,Female ,Trefoil Factor-1 ,Tyrosine kinase ,medicine.drug - Abstract
Treatment with tyrosine kinase inhibitors (TKIs) including trastuzumab has revolutionized the management of HER2-positive breast cancer. Recent evaluation of clinical trial data suggests that a subset of HER2/ER double-positive cancers may not receive significant benefit from the TKI therapy. Here we investigate the cross talk between HER2 and ER in breast cancer and monitor the effect of trastuzumab on the tyrosine kinase effector transcription factor Myc. In HER2-positive breast cancer patients treated with neoadjuvant trastuzumab, steroid receptor-negative status (ER and PR negative) of pre-treatment biopsies predicted pathological complete response (pCR) (n=31 patients, P=0.0486), whereas elevated Myc protein inversely associated with pCR (P=0.0446). Liquid chromatography mass spectrometry identified the corepressor SMRT as a novel Myc-interacting protein. Trastuzumab treatment enhanced Myc-SMRT interactions in HER2-overexpressing breast cancer cells (LCC1) and inhibited expression of the Myc target gene survivin. In HER2-low, ER-positive steroid-dominant cells (MCF7), trastuzumab therapy repressed Myc-SMRT interactions and upregulated survivin expression. Trastuzumab treatment induced ER-CBP interactions, enhanced ER transcriptional activity and upregulated expression of the ER target gene pS2. The absence of pS2 expression in pre-treatment biopsies predicted pCR to neoadjuvant trastuzumab in breast cancer patients (n=25, P=0.0089) and pS2 expression associated with residual cancer burden (P=0.0196). Furthermore, metastatic tissues from patients who had failed trastuzumab therapy were pS2 positive. In HER2-overexpressing cells, trastuzumab treatment can repress Myc transcriptional activity and clinical response is favorable. However, with co-expression of the steroid pathway, this inhibition is lost and response to treatment is often poor.
- Published
- 2014
- Full Text
- View/download PDF
19. Patient-Derived Xenografts of Breast Cancer
- Author
-
Damir, Varešlija, Sinead, Cocchiglia, Christopher, Byrne, and Leonie, Young
- Subjects
Disease Models, Animal ,Mice ,Carcinogenesis ,Animals ,Heterografts ,Humans ,Breast Neoplasms ,Female ,Xenograft Model Antitumor Assays - Abstract
With the advancement of translational research, particularly in the field of cancer, it is now imperative to have models which more clearly reflect patient heterogeneity. Patient derived xenograft (PDX) models, which involve the orthotopic implantation of breast tumors into immune-compromised mice, recapitulate the native tumor biology. Despite the considerable challenges that establishing PDX models present, they are the ultimate model to study tumorigenesis of refractory disease and for assessing the efficacy of new pharmaceutical compounds.
- Published
- 2016
20. Patient-Derived Xenografts of Breast Cancer
- Author
-
Christopher Byrne, Sinead Cocchiglia, Leonie S. Young, and Damir Varešlija
- Subjects
0301 basic medicine ,Tumor biology ,business.industry ,Refractory Disease ,Cancer ,Translational research ,medicine.disease ,medicine.disease_cause ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Cancer research ,Carcinogenesis ,business ,Tumor xenograft - Abstract
With the advancement of translational research, particularly in the field of cancer, it is now imperative to have models which more clearly reflect patient heterogeneity. Patient derived xenograft (PDX) models, which involve the orthotopic implantation of breast tumors into immune-compromised mice, recapitulate the native tumor biology. Despite the considerable challenges that establishing PDX models present, they are the ultimate model to study tumorigenesis of refractory disease and for assessing the efficacy of new pharmaceutical compounds.
- Published
- 2016
- Full Text
- View/download PDF
21. Abstract P5-04-12: Withdrawn
- Author
-
Damir Varešlija, Sinead Cocchiglia, Ben Doherty, Sara Charmsaz, Damian McCartan, L Young, Jarlath C. Bolger, and A Hill
- Subjects
Cancer Research ,Oncology - Abstract
This abstract was withdrawn by the authors. Citation Format: Charmsaz S, Doherty B, Cocchiglia S, Bolger J, Vareslija D, McCartan D, Hill A, Young L. Withdrawn [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P5-04-12.
- Published
- 2019
- Full Text
- View/download PDF
22. AB066. 208. Loss of estrogen receptor (ER) through DNA methylation and alterations in molecular heterogeneity during endocrine treatment in ER-positive breast cancer
- Author
-
Michael Flanagan, Nicola Cosgrove, Sinead Cocchiglia, Arnold K. Hill, Damir Varšlija, Sara Charmsaz, and L Young
- Subjects
DNA methylation ,Cancer research ,Estrogen receptor ,Endocrine system ,General Medicine ,Biology ,Molecular heterogeneity - Published
- 2019
- Full Text
- View/download PDF
23. PGC-1α is coupled to HIF-1α-dependent gene expression by increasing mitochondrial oxygen consumption in skeletal muscle cells
- Author
-
Sinead Cocchiglia, Eoin P. Cummins, Kathleen A. O'Hagan, Terence A. Agbor, John F. Garvey, Mona Monfared, Cormac T. Taylor, Alexander V. Zhdanov, Bernard B. Allan, Murtaza M. Tambuwala, and Dmitri B. Papkovsky
- Subjects
Peroxisome proliferator-activated receptor ,chemistry.chemical_element ,Mitochondrion ,Biology ,Oxygen ,Gene Expression Regulation, Enzymologic ,Mice ,Oxygen Consumption ,medicine ,Animals ,Humans ,Muscle, Skeletal ,Heat-Shock Proteins ,chemistry.chemical_classification ,Regulation of gene expression ,Multidisciplinary ,RNA-Binding Proteins ,Skeletal muscle ,Biological Sciences ,Peroxisome ,Hypoxia-Inducible Factor 1, alpha Subunit ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Mitochondria ,Rats ,Cell biology ,PPAR gamma ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Mitochondrial biogenesis ,Trans-Activators ,Protein stabilization ,Transcription Factors - Abstract
Mitochondrial biogenesis occurs in response to increased cellular ATP demand. The mitochondrial electron transport chain requires molecular oxygen to produce ATP. Thus, increased ATP generation after mitochondrial biogenesis results in increased oxygen demand that must be matched by a corresponding increase in oxygen supply. We found that overexpression of peroxisome proliferator-activated receptor-gamma coactivator 1alpha (PGC-1alpha), which increases mitochondrial biogenesis in primary skeletal muscle cells, leads to increased expression of a cohort of genes known to be regulated by the dimeric hypoxia-inducible factor (HIF), a master regulator of the adaptive response to hypoxia. PGC-1alpha-dependent induction of HIF target genes under physiologic oxygen concentrations is not through transcriptional coactivation of HIF or up-regulation of HIF-1alpha mRNA but through HIF-1alpha protein stabilization. It occurs because of intracellular hypoxia as a result of increased oxygen consumption after mitochondrial biogenesis. Thus, we propose that at physiologic oxygen concentrations, PGC-1alpha is coupled to HIF signaling through the regulation of intracellular oxygen availability, allowing cells and tissues to match increased oxygen demand after mitochondrial biogenesis with increased oxygen supply.
- Published
- 2009
- Full Text
- View/download PDF
24. Ets-2 and p160 proteins collaborate to regulate c-Myc in endocrine resistant breast cancer
- Author
-
Leonie S. Young, Gabrielle E. Kelly, Dhafir Al-Azawi, A Hill, M Mc Ilroy, Sinead Cocchiglia, Fiona T Bane, and Aisling M. Redmond
- Subjects
Nucleocytoplasmic Transport Proteins ,Cancer Research ,Antineoplastic Agents, Hormonal ,Breast Neoplasms ,Biology ,medicine.disease_cause ,DNA-binding protein ,Proto-Oncogene Protein c-ets-2 ,Proto-Oncogene Proteins c-myc ,Cell Line, Tumor ,Coactivator ,Genetics ,medicine ,Humans ,Molecular Biology ,Transcription factor ,Effector ,Nuclear Proteins ,RNA-Binding Proteins ,Cancer ,medicine.disease ,DNA-Binding Proteins ,Tamoxifen ,Cancer research ,Oncogene MYC ,Female ,Breast disease ,Carcinogenesis ,Protein Binding ,Transcription Factors - Abstract
Associations between p160 coactivator proteins and endocrine resistance have been described. Though thought to primarily interact with steroid receptors, the p160 proteins can also interact with non-nuclear receptor transcription factors including the MAP kinase effector proteins Ets. Here, we observed that in breast cancer cells resistant and insensitive to endocrine treatment, the growth factor EGF induced Ets-2 but not Ets-1 transcriptional regulation of the oncogene myc. Ets-2 regulation of myc was found to be reliant on the p160 proteins SRC-1 and SRC-3. In support of these molecular observations, strong associations were observed between the transcription factor, Ets-2 and its coactivator SRC-1 (P
- Published
- 2007
- Full Text
- View/download PDF
25. Mapping molecular subtype specific alterations in breast cancer brain metastases identifies clinically relevant vulnerabilities
- Author
-
Nicola Cosgrove, Damir Varešlija, Stephen Keelan, Ashuvinee Elangovan, Jennifer M. Atkinson, Sinéad Cocchiglia, Fiona T. Bane, Vikrant Singh, Simon Furney, Chunling Hu, Jodi M. Carter, Steven N. Hart, Siddhartha Yadav, Matthew P. Goetz, Arnold D. K. Hill, Steffi Oesterreich, Adrian V. Lee, Fergus J. Couch, and Leonie S. Young
- Subjects
Science - Abstract
The molecular landscape of breast cancer brain metastases (BCBM) is still understudied, especially for different breast cancer subtypes. Here, the authors characterise subtype-specific BCBMs using genomics and transcriptomics and identify homologous recombination deficiency as a key therapeutic vulnerability.
- Published
- 2022
- Full Text
- View/download PDF
26. Metastatic progression with resistance to aromatase inhibitors is driven by the steroid receptor coactivator SRC-1
- Author
-
Sarah M. Theissen, Stephen Sande, E Hughes, Leonie S. Young, Arnold D.K. Hill, Christopher Byrne, Paul Tibbitts, Jean McBryan, Jane O'Hara, and Sinead Cocchiglia
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Breast Neoplasms ,Transfection ,Breast cancer ,Nuclear Receptor Coactivator 1 ,Internal medicine ,Cell Line, Tumor ,Coactivator ,medicine ,Humans ,Aromatase ,Neoplasm Metastasis ,Receptor ,biology ,business.industry ,Aromatase Inhibitors ,Letrozole ,medicine.disease ,Primary tumor ,Tamoxifen ,Endocrinology ,Oncology ,Hormone receptor ,Estrogen ,biology.protein ,Cancer research ,Disease Progression ,Female ,business ,medicine.drug - Abstract
Aromatase inhibitors (AI) are a standard-of-care treatment for postmenopausal, estrogen receptor–positive breast cancers. Although tumor recurrence on AI therapy occurs, the mechanisms underlying acquired resistance to AIs remain unknown. In this study, we examined a cohort of endocrine-treated breast cancer patients and used a cell line model of resistance to the AI letrozole. In patients treated with a first-line AI, hormone receptor switching between primary and resistant tumors was a common feature of disease recurrence. Resistant cells exhibited a switch from steroid-responsive growth to growth factor–responsive and endocrine-independent growth, which was accompanied by the development of a more migratory and disorganized phenotype. Both the resistant cells and tumors from AI-resistant patients showed high expression of the steroid receptor coactivator SRC-1. Direct interactions between SRC-1 and the transcription factor Ets2 regulated Myc and MMP9. SRC-1 was required for the aggressive and motile phenotype of AI-resistant cells. Interestingly, SRC-1 expression in primary and/or recurrent tumors was associated with a reduction in disease-free survival in treated patients. Moreover, there was a significant association between SRC-1 and Ets2 in the recurrent tissue compared with the matched primary tumor. Together, our findings elucidate a mechanism of AI-specific metastatic progression in which interactions between SRC-1 and Ets2 promote dedifferentiation and migration in hormone-dependent breast cancer. Cancer Res; 72(2); 548–59. ©2011 AACR.
- Published
- 2011
27. ADAM22/LGI1 complex as a new actionable target for breast cancer brain metastasis
- Author
-
Sara Charmsaz, Ben Doherty, Sinéad Cocchiglia, Damir Varešlija, Attilio Marino, Nicola Cosgrove, Ricardo Marques, Nolan Priedigkeit, Siobhan Purcell, Fiona Bane, Jarlath Bolger, Christopher Byrne, Philip J. O’Halloran, Francesca Brett, Katherine Sheehan, Kieran Brennan, Ann M. Hopkins, Stephen Keelan, Petra Jagust, Stephen Madden, Chiara Martinelli, Matteo Battaglini, Steffi Oesterreich, Adrian V. Lee, Gianni Ciofani, Arnold D. K. Hill, and Leonie S. Young
- Subjects
Breast cancer metastases ,Brain metastases ,ADAM22 ,LGI1 ,ECM signalling ,Blood–brain barrier ,Medicine - Abstract
Abstract Background Metastatic breast cancer is a major cause of cancer-related deaths in woman. Brain metastasis is a common and devastating site of relapse for several breast cancer molecular subtypes, including oestrogen receptor-positive disease, with life expectancy of less than a year. While efforts have been devoted to developing therapeutics for extra-cranial metastasis, drug penetration of blood–brain barrier (BBB) remains a major clinical challenge. Defining molecular alterations in breast cancer brain metastasis enables the identification of novel actionable targets. Methods Global transcriptomic analysis of matched primary and metastatic patient tumours (n = 35 patients, 70 tumour samples) identified a putative new actionable target for advanced breast cancer which was further validated in vivo and in breast cancer patient tumour tissue (n = 843 patients). A peptide mimetic of the target’s natural ligand was designed in silico and its efficacy assessed in in vitro, ex vivo and in vivo models of breast cancer metastasis. Results Bioinformatic analysis of over-represented pathways in metastatic breast cancer identified ADAM22 as a top ranked member of the ECM-related druggable genome specific to brain metastases. ADAM22 was validated as an actionable target in in vitro, ex vivo and in patient tumour tissue (n = 843 patients). A peptide mimetic of the ADAM22 ligand LGI1, LGI1MIM, was designed in silico. The efficacy of LGI1MIM and its ability to penetrate the BBB were assessed in vitro, ex vivo and in brain metastasis BBB 3D biometric biohybrid models, respectively. Treatment with LGI1MIM in vivo inhibited disease progression, in particular the development of brain metastasis. Conclusion ADAM22 expression in advanced breast cancer supports development of breast cancer brain metastasis. Targeting ADAM22 with a peptide mimetic LGI1MIM represents a new therapeutic option to treat metastatic brain disease.
- Published
- 2020
- Full Text
- View/download PDF
28. Protein macroarray identification of biomarkers in HER2-positive breast cancer
- Author
-
Sinead Cocchiglia, Arnold D.K. Hill, Derek Murphy, D. Collins, Leonie S. Young, and Hong Chen
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,fungi ,food and beverages ,medicine.disease ,Response to treatment ,Holy Grail ,Breast cancer ,Internal medicine ,HER2 Positive Breast Cancer ,medicine ,Identification (biology) ,business - Abstract
e11575 Background: The holy grail of breast cancer diagnosis and management is the identification of biomarkers that can simplify diagnosis, predict response to treatment and facilitate follow-up t...
- Published
- 2014
- Full Text
- View/download PDF
29. RuvBl2 cooperates with Ets2 to transcriptionally regulate hTERT in colon cancer
- Author
-
Sinead Cocchiglia, Arnold D.K. Hill, Patrick Fahy-Browne, Jean McBryan, Elaine W. Kay, Kilian Perrem, Paul Tibbitts, Aisling M. Redmond, Achim Treumann, Leonie S. Young, Marie McIlroy, and Padraic Flavin
- Subjects
Senescence ,Proteomics ,Telomerase ,Transcription, Genetic ,Colorectal cancer ,Cell ,Biophysics ,Biology ,Biochemistry ,Proto-Oncogene Protein c-ets-2 ,RuvBl2 ,Structural Biology ,Genetics ,RUVBL2 ,Transcriptional regulation ,medicine ,Humans ,Telomerase reverse transcriptase ,Molecular Biology ,DNA Helicases ,Cell Biology ,medicine.disease ,Molecular biology ,Telomere ,Colon cancer ,Gene Expression Regulation, Neoplastic ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,c-Myc ,Colonic Neoplasms ,Cancer research ,Disease Progression ,ATPases Associated with Diverse Cellular Activities ,Carrier Proteins ,hTERT - Abstract
Human cancers utilise telomerase to maintain telomeres and prohibit cell senescence. Human telomerase reverse transcriptase (hTERT), an essential component of this complex, is regulated at the level of gene transcription. Using SILAC-proteomic analysis and molecular studies, we identified the AAA+ ATPase, RuvBl2 as a transcriptional regulator of hTERT and established that this regulation is through cooperation with Ets-2. In colon cancer patients, nuclear expression of RuvBl2 associated with nuclear expression of hTERT, pEts2 and advanced nodal disease (P < 0.01, P = 0.05 and P = 0.03 respectively, n = 170). These data firmly implicate RuvBl2 in Ets2 mediated regulation of hTERT in colon cancer which has functional and clinical consequences.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.