Nikiana Simigdala
Institute of Cancer Research
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Nikiana Simigdala.
Breast Cancer Research | 2016
Nikiana Simigdala; Qiong Gao; Sunil Pancholi; Hanne Roberg-Larsen; Marketa Zvelebil; Ricardo Ribas; Elizabeth Folkerd; Andrew Thompson; Amandeep Bhamra; Mitch Dowsett; Lesley-Ann Martin
BackgroundTherapies targeting estrogenic stimulation in estrogen receptor-positive (ER+) breast cancer (BC) reduce mortality, but resistance remains a major clinical problem. Molecular studies have shown few high-frequency mutations to be associated with endocrine resistance. In contrast, expression profiling of primary ER+ BC samples has identified several promising signatures/networks for targeting.MethodsTo identify common adaptive mechanisms associated with resistance to aromatase inhibitors (AIs), we assessed changes in global gene expression during adaptation to long-term estrogen deprivation (LTED) in a panel of ER+ BC cell lines cultured in 2D on plastic (MCF7, T47D, HCC1428, SUM44 and ZR75.1) or in 3D on collagen (MCF7) to model the stromal compartment. Furthermore, dimethyl labelling followed by LC-MS/MS was used to assess global changes in protein abundance. The role of target genes/proteins on proliferation, ER-mediated transcription and recruitment of ER to target gene promoters was analysed.ResultsThe cholesterol biosynthesis pathway was the common upregulated pathway in the ER+ LTED but not the ER– LTED cell lines, suggesting a potential mechanism dependent on continued ER expression. Targeting the individual genes of the cholesterol biosynthesis pathway with siRNAs caused a 30–50 % drop in proliferation. Further analysis showed increased expression of 25-hydroxycholesterol (HC) in the MCF7 LTED cells. Exogenous 25-HC or 27-HC increased ER-mediated transcription and expression of the endogenous estrogen-regulated gene TFF1 in ER+ LTED cells but not in the ER– LTED cells. Additionally, recruitment of the ER and CREB-binding protein (CBP) to the TFF1 and GREB1 promoters was increased upon treatment with 25-HC and 27-HC. In-silico analysis of two independent studies of primary ER+ BC patients treated with neoadjuvant AIs showed that increased expression of MSMO1, EBP, LBR and SQLE enzymes, required for cholesterol synthesis and increased in our in-vitro models, was significantly associated with poor response to endocrine therapy.ConclusionTaken together, these data provide support for the role of cholesterol biosynthesis enzymes and the cholesterol metabolites, 25-HC and 27-HC, in a novel mechanism of resistance to endocrine therapy in ER+ BC that has potential as a therapeutic target.
Molecular Cancer Therapeutics | 2015
Ricardo Ribas; Sunil Pancholi; Stephanie K. Guest; Elisabetta Marangoni; Qiong Gao; Aurélie Thuleau; Nikiana Simigdala; Urszula M. Polanska; Hayley Campbell; Aradhana Rani; Gianmaria Liccardi; Stephen R. D. Johnston; Barry R. Davies; Mitch Dowsett; Lesley-Ann Martin
PI3K/AKT/mTOR signaling plays an important role in breast cancer. Its interaction with estrogen receptor (ER) signaling becomes more complex and interdependent with acquired endocrine resistance. Targeting mTOR combined with endocrine therapy has shown clinical utility; however, a negative feedback loop exists downstream of PI3K/AKT/mTOR. Direct blockade of AKT together with endocrine therapy may improve breast cancer treatment. AZD5363, a novel pan-AKT kinase catalytic inhibitor, was examined in a panel of ER+ breast cancer cell lines (MCF7, HCC1428, T47D, ZR75.1) adapted to long-term estrogen deprivation (LTED) or tamoxifen (TamR). AZD5363 caused a dose-dependent decrease in proliferation in all cell lines tested (GI50 < 500 nmol/L) except HCC1428 and HCC1428-LTED. T47D-LTED and ZR75-LTED were the most sensitive of the lines (GI50 ∼100 nmol/L). AZD5363 resensitized TamR cells to tamoxifen and acted synergistically with fulvestrant. AZD5363 decreased p-AKT/mTOR targets leading to a reduction in ERα-mediated transcription in a context-specific manner and concomitant decrease in recruitment of ER and CREB-binding protein (CBP) to estrogen response elements located on the TFF1, PGR, and GREB1 promoters. Furthermore, AZD5363 reduced expression of cell-cycle–regulatory proteins. Global gene expression highlighted ERBB2-ERBB3, ERK5, and IGFI signaling pathways driven by MYC as potential feedback-loops. Combined treatment with AZD5363 and fulvestrant showed synergy in an ER+ patient-derived xenograft and delayed tumor progression after cessation of therapy. These data support the combination of AZD5363 with fulvestrant as a potential therapy for breast cancer that is sensitive or resistant to E-deprivation or tamoxifen and that activated AKT is a determinant of response, supporting the need for clinical evaluation. Mol Cancer Ther; 14(9); 2035–48. ©2015 AACR.
Nature Communications | 2017
Lesley-Ann Martin; Ricardo Ribas; Nikiana Simigdala; Eugene Schuster; Sunil Pancholi; Tencho Tenev; Pascal Gellert; Laki Buluwela; Alison Harrod; Allan Thornhill; Joanna Nikitorowicz-Buniak; Amandeep Bhamra; Marc-Olivier Turgeon; George Poulogiannis; Qiong Gao; Vera Martins; Margaret Hills; Isaac Garcia-Murillas; Charlotte Fribbens; Neill Patani; Zheqi Li; Matthew J. Sikora; Nicholas C. Turner; Wilbert Zwart; Steffi Oesterreich; Jason S. Carroll; Simak Ali; Mitch Dowsett
Resistance to endocrine therapy remains a major clinical problem in breast cancer. Genetic studies highlight the potential role of estrogen receptor-α (ESR1) mutations, which show increased prevalence in the metastatic, endocrine-resistant setting. No naturally occurring ESR1 mutations have been reported in in vitro models of BC either before or after the acquisition of endocrine resistance making functional consequences difficult to study. We report the first discovery of naturally occurring ESR1Y537C and ESR1Y537S mutations in MCF7 and SUM44 ESR1-positive cell lines after acquisition of resistance to long-term-estrogen-deprivation (LTED) and subsequent resistance to fulvestrant (ICIR). Mutations were enriched with time, impacted on ESR1 binding to the genome and altered the ESR1 interactome. The results highlight the importance and functional consequence of these mutations and provide an important resource for studying endocrine resistance.ESR1 mutations occur in endocrine-resistant patients but have not yet been reported in in vitro models of breast cancer. Here, the authors report the discovery of naturally occurring ESR1Y537Cand ESR1Y537S mutations in two breast cancer cell lines after acquisition of resistance to long-term-estrogen-deprivation.
PLOS ONE | 2016
Stephanie K. Guest; Ricardo Ribas; Sunil Pancholi; Joanna Nikitorowicz-Buniak; Nikiana Simigdala; Mitch Dowsett; Stephen R. D. Johnston; Lesley-Ann Martin
Despite the effectiveness of endocrine therapies in estrogen receptor positive (ER+) breast cancer, approximately 40% of patients relapse. Previously, we identified the Focal-adhesion kinase canonical pathway as a major contributor of resistance to estrogen deprivation and cellular-sarcoma kinase (c-src) as a dominant gene in this pathway. Dasatinib, a pan-src inhibitor, has recently been used in clinical trials to treat ER+ patients but has shown mixed success. In the following study, using isogenic cell line models, we provide a potential explanation for these findings and suggest a sub-group that may benefit. A panel of isogenic cell lines modelling resistance to aromatase inhibitors (LTED) and tamoxifen (TAMR) were assessed for response to dasatinib ± endocrine therapy. Dasatinib caused a dose-dependent decrease in proliferation in MCF7-TAMR cells and resensitized them to tamoxifen and fulvestrant but not in HCC1428-TAMR. In contrast, in estrogen-deprived conditions, dasatinib increased the proliferation rate of parental-MCF7 cells and had no effect on MCF7-LTED or HCC1428-LTED. Treatment with dasatinib caused a decrease in src-phosphorylation and inhibition of downstream pathways, including AKT and ERK1/2 in all cell lines tested, but only the MCF7-TAMR showed a concomitant decrease in markers of cell cycle progression. Inhibition of src also caused a significant decrease in cell migration in both MCF7-LTED and MCF7-TAMR cells. Finally, we showed that, in MCF7-TAMR cells, in contrast to tamoxifen sensitive cell lines, ER is expressed throughout the cell rather than being restricted to the nucleus and that treatment with dasatinib resulted in nuclear shuttling of ER, which was associated with an increase in ER-mediated transcription. These data suggest that src has differential effects in endocrine-resistant cell lines, particularly in tamoxifen resistant models, with low ER genomic activity, providing further evidence of the importance of patient selection for clinical trials testing dasatinib utility in ER+ breast cancer.
Nature Communications | 2018
Joseph S. Baxter; Olivia C. Leavy; Nicola Dryden; Sarah Maguire; Nichola Johnson; Vita Fedele; Nikiana Simigdala; Lesley-Ann Martin; Simon Andrews; Steven W. Wingett; Ioannis Assiotis; Kerry Fenwick; Ritika Chauhan; Alistair G. Rust; Nick Orr; Frank Dudbridge; Syed Haider; Olivia Fletcher
Genome-wide association studies (GWAS) have identified approximately 100 breast cancer risk loci. Translating these findings into a greater understanding of the mechanisms that influence disease risk requires identification of the genes or non-coding RNAs that mediate these associations. Here, we use Capture Hi-C (CHi-C) to annotate 63 loci; we identify 110 putative target genes at 33 loci. To assess the support for these target genes in other data sources we test for associations between levels of expression and SNP genotype (eQTLs), disease-specific survival (DSS), and compare them with somatically mutated cancer genes. 22 putative target genes are eQTLs, 32 are associated with DSS and 14 are somatically mutated in breast, or other, cancers. Identifying the target genes at GWAS risk loci will lead to a greater understanding of the mechanisms that influence breast cancer risk and prognosis.Risk loci for breast cancer have been identified by genome-wide association studies. Here, the authors use Capture Hi-C to identify 110 putative target genes at 33 loci and assessed associations of gene expression, SNP genotype, and survival, providing evidence of mechanisms that may influence the prognosis and risk of breast cancer.
British Journal of Cancer | 2018
Nikiana Simigdala; Sunil Pancholi; Ricardo Ribas; Elizabeth Folkerd; Gianmaria Liccardi; Joanna Nikitorowicz-Buniak; Stephen R. D. Johnston; Mitch Dowsett; Lesley-Ann Martin
BackgroundResistance to endocrine therapy remains a major clinical problem in the treatment of oestrogen-receptor positive (ER+) breast cancer. Studies show androgen-receptor (AR) remains present in 80–90% of metastatic breast cancers providing support for blockade of AR-signalling. However, clinical studies with abiraterone, which blocks cytochrome P450 17A1 (CYP17A1) showed limited benefit.MethodsIn order to address this, we assessed the impact of abiraterone on cell-viability, cell-death, ER-mediated transactivation and recruitment to target promoters. together with ligand-binding assays in a panel of ER+ breast cancer cell lines that were either oestrogen-dependent, modelling endocrine-sensitive disease, or oestrogen-independent modelling relapse on an aromatase inhibitor. The latter, harboured wild-type (wt) or naturally occurring ESR1 mutations.ResultsSimilar to oestrogen, abiraterone showed paradoxical impact on proliferation by stimulating cell growth or death, depending on whether the cells are hormone-dependent or have undergone prolonged oestrogen-deprivation, respectively. Abiraterone increased ER-turnover, induced ER-mediated transactivation and ER-degradation via the proteasome.ConclusionsOur study confirms the oestrogenic activity of abiraterone and highlights its differential impact on cells dependent on oestrogen for their proliferation vs. those that are ligand-independent and harbour wt or mutant ESR1. These properties could impact the clinical efficacy of abiraterone in breast cancer.
Breast Cancer Research | 2018
Ricardo Ribas; Sunil Pancholi; Aradhana Rani; Eugene Schuster; Stephanie K. Guest; Joanna Nikitorowicz-Buniak; Nikiana Simigdala; Allan Thornhill; Francesca Avogadri-Connors; Richard E. Cutler; Alshad S. Lalani; Mitch Dowsett; Stephen R. D. Johnston; Lesley-Ann Martin
BackgroundEndocrine therapies are the mainstay of treatment for oestrogen receptor (ER)-positive (ER+) breast cancer (BC). However, resistance remains problematic largely due to enhanced cross-talk between ER and growth factor pathways, circumventing the need for steroid hormones. Previously, we reported the anti-proliferative effect of everolimus (RAD001-mTORC1 inhibitor) with endocrine therapy in resistance models; however, potential routes of escape from treatment via ERBB2/3 signalling were observed. We hypothesised that combined targeting of three cellular nodes (ER, ERBB, and mTORC1) may provide enhanced long-term clinical utility.MethodsA panel of ER+ BC cell lines adapted to long-term oestrogen deprivation (LTED) and expressing ESR1wt or ESR1Y537S, modelling acquired resistance to an aromatase-inhibitor (AI), were treated in vitro with a combination of RAD001 and neratinib (pan-ERBB inhibitor) in the presence or absence of oestradiol (E2), tamoxifen (4-OHT), or fulvestrant (ICI182780). End points included proliferation, cell signalling, cell cycle, and effect on ER-mediated transactivation. An in-vivo model of AI resistance was treated with monotherapies and combinations to assess the efficacy in delaying tumour progression. RNA-seq analysis was performed to identify changes in global gene expression as a result of the indicated therapies.ResultsHere, we show RAD001 and neratinib (pan-ERBB inhibitor) caused a concentration-dependent decrease in proliferation, irrespective of the ESR1 mutation status. The combination of either agent with endocrine therapy further reduced proliferation but the maximum effect was observed with a triple combination of RAD001, neratinib, and endocrine therapy. In the absence of oestrogen, RAD001 caused a reduction in ER-mediated transcription in the majority of the cell lines, which associated with a decrease in recruitment of ER to an oestrogen-response element on the TFF1 promoter. Contrastingly, neratinib increased both ER-mediated transactivation and ER recruitment, an effect reduced by the addition of RAD001. In-vivo analysis of an LTED model showed the triple combination of RAD001, neratinib, and fulvestrant was most effective at reducing tumour volume. Gene set enrichment analysis revealed that the addition of neratinib negated the epidermal growth factor (EGF)/EGF receptor feedback loops associated with RAD001.ConclusionsOur data support the combination of therapies targeting ERBB2/3 and mTORC1 signalling, together with fulvestrant, in patients who relapse on endocrine therapy and retain a functional ER.
Molecular Cancer Research | 2016
Nikiana Simigdala; Qiong Gao; Sunil Pancholi; Marketa Zvelebil; Ricardo Ribas; Mitch Dowsett; Lesley-Ann Martin
Over 80% of breast cancers (BC) at primary diagnosis express the estrogen receptor (ER+). Therapies targeting the estrogenic stimulation of tumor growth reduce mortality from ER+ BC but resistance remains a major clinical problem. Data from large studies such as TCGA indicate that other than a small number of high frequency mutations such as TP53, PIK3CA and GATA3 that have little association with endocrine resistance, primary ER+ BC shows very low frequency of individual mutations making targeting difficult. In contrast, expression profiling of primary ER+ BC samples have identified several promising signatures/networks for targeting. In order to identify common adaptive mechanisms associated with resistance to aromatase inhibitors, we assessed changes in global gene expression during adaptation to long-term estrogen deprivation (LTED) in a panel of ER+ BC cell lines cultured in 2D on plastic (MCF7, T47D, HCC1428, SUM44, MDA-MB-361 and ZR75.1) or 3D on collagen to model the stromal compartment (MCF7). At the point of resistance MCF7-LTED, HCC1428-LTED and SUM44-LTED retained ER expression whilst T47D-LTED, ZR75.1-LTED and MDA-MB-361-LTED reduced or lost ER. In order to identify common adaptive mechanisms, ingenuity pathway was used. Genes submitted for analysis were selected on the basis of a p-value of 0.001, FDR 5% and fold change 1.5 relative to the parental cell lines. Strikingly, the cholesterol biosynthesis pathway was the common up-regulated pathway in the ER+ LTED cell lines but not in the ER- LTED cell lines, suggesting a potential mechanism dependent on continued ER expression. Further interrogation showed increased expression of MSMO1 (x2.27), EBP (x1.57), SQLE (x2.37) and IDI1 (x2.74) in the MCF7-LTED. As mRNA does not always correlate with protein, we applied quantitative proteomics in wt-MCF7 and MCF7-LTED cells using dimethyl labelling followed by LC-MS/MS. Several enzymes within the cholesterol biosynthesis pathway were higher in abundance in the MCF7-LTED such as EBP (x1.54), SQLE (x3.24) and LBR (x1.86). Evidence suggests that patients with cancer accumulate cholesteryl esters as a result of loss of PTEN or PI3K/AKT/mTOR activation, stimulating SREBP, ACAT1, HMGSCR and LDLR. As 40% of ER+ BC harbor PTEN loss or PIK3CA mutations, we assessed levels of SREBP, HMGCR, LDLR or ACAT1 in our data sets. Neither SREBP nor HMGCR mRNA or protein levels changed in the ER+ LTED or ER- LTED. Furthermore, ACAT1 was downregulated in the ER+ LTED. Moreover, levels of free and esterified cholesterol in wt-MCF7 and MCF7-LTED cell lysates were unchanged. Previous studies have suggested oxysterols can act as selective ER-modulators (SERM). We subsequently investigated the function of these cholesterol metabolites in our LTED models. Treatment with 25-hydroxycholesterol (HC) or 27-HC increased ER mediated-transcription and expression of the endogenous estrogen-regulated gene TFF1 in ER+ LTED cells but not in the ER- LTED. Targeting the individual genes of the cholesterol biosynthesis pathway with siRNAs caused a c. 30-60% drop in proliferation and concomitant drop in TFF1 expression in the ER+ LTED cells together with a reduction in recruitment of the ER and CREB binding protein (CBP) to the TFF1 promoter. Gene expression data from 704 primary ER+ BC from patients treated with adjuvant tamoxifen showed that MSMO1 (p=0.047), EBP (p=0.043), SQLE (p=0.0001), DHCR7 (p=0.002) and IDI1 (p=0.0005), enzymes required for cholesterol synthesis and up-regulated in our in-vitro models, were associated with poor relapse free survival. Of note, DHCR7 forms part of the EndoPredict gene signature. Taken together, these data provide support for the role of the cholesterol biosynthesis pathway and its metabolites as a novel mechanism of resistance to endocrine therapy in ER+ BC and its potential as a therapeutic target. Citation Format: Nikiana Simigdala, Qiong Gao, Sunil Pancholi, Marketa Zvelebil, Ricardo Ribas, Mitch Dowsett, Lesley-Ann Martin. Cholesterol biosynthesis pathway as a novel mechanism of resistance identified in ER+ long-term estrogen deprived cells. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research; Oct 17-20, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(2_Suppl):Abstract nr A37.
Cancer Research | 2015
Lesley-Ann Martin; Stephanie K. Guest; Sunil Pancholi; Ricardo Ribas; Qiong Gao; Nikiana Simigdala; Aradhana Rani; Barry R. Davies; Stephen R. D. Johnston; Mitch Dowsett
AIM: To evaluate the efficacy and functional consquences of combining AZD5363 with endocrine therapy in pre-clinical models of endocrine-sensitive and resistant ER+ BC. BACKGROUND: The PI3K/AKT/mTOR signalling pathway plays an important role in BC. Its close interaction with ER signalling becomes more complex and inter-dependent with acquisition of endocrine resistance. Targeting the mTOR pathway in combination with endocrine therapy has shown clinical utility. However, a negative feedback loop exists downstream of the PI3K/AKT/mTOR pathway with mTOR inhibition leading to increased activation of IGFR1-dependent AKT activity potentially negating long-term benefit. Direct blockade of AKT in combination with endocrine therapy, may provide a better rationale for treatment of endocrine-resistant BC, impacting on both cell survival/apoptosis and ER ligand-independent signaling. In this investigation, we assessed the efficacy of AZD5363, a pan-AKT inhibitor with endocrine therapies in pre-clinical models of endocrine sensitive and resistant ER+ BC and its impact on molecular and cellular response. METHODS: Inhibition of AKT using AZD5363 was examined in 5 ER+ BC lines before and after adaptation to long-term estrogen deprivation (LTED) or tamoxifen (TAMR). The effects of AZD5363 on cell proliferation were determined alone and in combination with endocrine treatment and feedback upregulation and activation of receptor tyrosine kinases (RTKs) was examined by western blotting. ER-transactivation was measured with an estrogen-response element (ERE)-linked luciferase reporter construct and confirmed using chromatin-immunoprecipitation. Global gene expression analysis was used to identify pathways associated with response. Xenografts were treated with AZD5363 ± fulvestrant to determine in vivo effects. RESULTS: AZD5363 caused a dose-dependent decrease in proliferation in all cell lines tested (GI50 CONCLUSION: These data suggest that AZD5363 plus fulvestrant may be effective in BC that is sensitive or resistant to E-deprivation or tamoxifen and that activated AKT is a determinant of response. These data strongly support the need for clinical evaluation. Citation Format: Lesley-Ann Martin, Stephanie K Guest, Sunil Pancholi, Ricardo Ribas, Qiong Gao, Nikiana Simigdala, Aradhana Rani, Barry Davies, Stephen Johnston, Mitch Dowsett. AKT antagonist AZD5363 targets estrogen receptor (ER) function in endocrine resistant breast cancer (BC) and synergises with fulvestrant in vivo [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P3-05-04.
Cancer Research | 2018
Joanna Nikitorowicz-Buniak; Sunil Pancholi; Nikiana Simigdala; Ricardo Ribas; Spiros Linardopoulos; Mitch Dowsett; Stephen Albert Johnston; Lesley-Ann Martin