Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where William Court is active.

Publication


Featured researches published by William Court.


BMC Biology | 2012

Advances in establishment and analysis of three-dimensional tumor spheroid-based functional assays for target validation and drug evaluation

Maria Vinci; Sharon Gowan; Frances E. Boxall; Lisa Patterson; Miriam Zimmermann; William Court; Cara Lomas; Marta Mendiola; David Hardisson; Suzanne A. Eccles

BackgroundThere is overwhelming evidence that in vitro three-dimensional tumor cell cultures more accurately reflect the complex in vivo microenvironment than simple two-dimensional cell monolayers, not least with respect to gene expression profiles, signaling pathway activity and drug sensitivity. However, most currently available three-dimensional techniques are time consuming and/or lack reproducibility; thus standardized and rapid protocols are urgently needed.ResultsTo address this requirement, we have developed a versatile toolkit of reproducible three-dimensional tumor spheroid models for dynamic, automated, quantitative imaging and analysis that are compatible with routine high-throughput preclinical studies. Not only do these microplate methods measure three-dimensional tumor growth, but they have also been significantly enhanced to facilitate a range of functional assays exemplifying additional key hallmarks of cancer, namely cell motility and matrix invasion. Moreover, mutual tissue invasion and angiogenesis is accommodated by coculturing tumor spheroids with murine embryoid bodies within which angiogenic differentiation occurs. Highly malignant human tumor cells were selected to exemplify therapeutic effects of three specific molecularly-targeted agents: PI-103 (phosphatidylinositol-3-kinase (PI3K)-mammalian target of rapamycin (mTOR) inhibitor), 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) (heat shock protein 90 (HSP90) inhibitor) and CCT130234 (in-house phospholipase C (PLC)γ inhibitor). Fully automated analysis using a Celigo cytometer was validated for tumor spheroid growth and invasion against standard image analysis techniques, with excellent reproducibility and significantly increased throughput. In addition, we discovered key differential sensitivities to targeted agents between two-dimensional and three-dimensional cultures, and also demonstrated enhanced potency of some agents against cell migration/invasion compared with proliferation, suggesting their preferential utility in metastatic disease.ConclusionsWe have established and validated a suite of highly reproducible tumor microplate three-dimensional functional assays to enhance the biological relevance of early preclinical cancer studies. We believe these assays will increase the translational predictive value of in vitro drug evaluation studies and reduce the need for in vivo studies by more effective triaging of compounds.


International Journal of Cancer | 2000

Overexpression of epidermal growth factor receptor in human head and neck squamous carcinoma cell lines correlates with matrix metalloproteinase‐9 expression and in vitro invasion

Pornchai O-charoenrat; Peter Rhys-Evans; Helmout Modjtahedi; William Court; Gary Box; Suzanne A. Eccles

Numerous reports have shown an association between overexpression of the epidermal growth factor receptor (EGFR), and poor prognosis in head and neck squamous cell carcinomas (HNSCC), however, the underlying mechanisms are still unclear. In the present study, we set out to determine whether EGFR expression was associated with in vitro invasive capacity in a panel of four established and ten newly derived HNSCC lines. Ten of the cell lines expressed high levels of EGFR as determined by a ligand‐binding assay and dot blot analysis, whereas the remaining four showed weak overexpression or normal levels of EGFR. The ability of cells to invade through Matrigel was found to be higher in the EGFR overexpressing cell lines (p < 0.0001). Expression levels of matrix metalloproteinases (MMP‐1, MMP‐2, MMP‐3, MMP‐7, MMP‐9, MMP‐10, MMP‐11, MMP‐13, MT1‐MMP) and tissue inhibitors of MMP (TIMP‐1, TIMP‐2) were evaluated by semiquantitative RT‐PCR, substrate zymography and western blot. We found a strong positive correlation between EGFR levels and the expression of MMP‐9 mRNA (r2 = 0.95; p < 0.0001), MMP‐9 enzyme activity (r2 = 0.8099; p < 0.0001) and an inverse correlation with TIMP‐1 (r2 = 0.48; p = 0.0059). In six selected HNSCC lines, in vitro invasion was assayed in the presence of an anti‐EGFR monoclonal antibody, ICR62. A significant reduction of invasion in four selected EGFR‐overexpressing cell lines was found with 30 nM ICR62 (from 50% to 70%; p < 0.001) but there was no effect in two cell lines with normal EGFR levels. Our results show that the in vitro invasive phenotype of HNSCC lines correlates with high EGFR and MMP‐9 expression, and it is therefore suggested that the EGFR signaling pathway might play an important role in the invasive behavior of HNSCC via specific upregulation of MMP‐9 and downregulation of TIMP‐1. Int. J. Cancer 86:307–317, 2000.


Biotechnology Annual Review | 2005

Cell migration/invasion assays and their application in cancer drug discovery

Suzanne A. Eccles; Carol Box; William Court

Invasive capacity is the single most important trait that distinguishes benign from malignant lesions. Tumour cells, during intravasation and extravasation of blood and lymphatic channels and when establishing colonies at secondary sites, must move through tissue boundaries that normal adult cells (other than, for example activated leukocytes) do not cross. Similar mechanisms are also utilised by activated endothelial cells during the generation of new blood vessels that enable the sustained growth and dissemination of tumours. It is now increasingly recognised that these processes--cell motility and invasion--might provide a rich source of novel targets for cancer therapy and that appropriate inhibitors may restrain both metastasis and neoangiogenesis. This new paradigm demands screening assays that can rapidly and quantitatively measure cell movement and the ability to traverse physiological barriers. We also need to consider whether simple reductionist in vitro approaches can reliably model the complexity of in vivo tumour invasion/neoangiogenesis. There are both opportunities and challenges ahead in developing a balanced portfolio of assays that will be able to evaluate accurately and finally deliver novel anti-invasive agents with therapeutic potential for clinical use.


Journal of the American Chemical Society | 2013

A Cyclic Peptide Inhibitor of HIF‑1 Heterodimerization That Inhibits Hypoxia Signaling in Cancer Cells

Elena Miranda; Ida Karin Nordgren; Abigail L. Male; Charlotte E. Lawrence; Franciane Hoakwie; Francesco Cuda; William Court; Keith R. Fox; Paul A. Townsend; Graham Packham; Suzanne A. Eccles; Ali Tavassoli

Hypoxia inducible factor-1 (HIF-1) is a heterodimeric transcription factor that acts as the master regulator of cellular response to reduced oxygen levels, thus playing a key role in the adaptation, survival, and progression of tumors. Here we report cyclo-CLLFVY, identified from a library of 3.2 million cyclic hexapeptides using a genetically encoded high-throughput screening platform, as an inhibitor of the HIF-1α/HIF-1β protein–protein interaction in vitro and in cells. The identified compound inhibits HIF-1 dimerization and transcription activity by binding to the PAS-B domain of HIF-1α, reducing HIF-1-mediated hypoxia response signaling in a variety of cell lines, without affecting the function of the closely related HIF-2 isoform. The reported cyclic peptide demonstrates the utility of our high-throughput screening platform for the identification of protein–protein interaction inhibitors, and forms the starting point for the development of HIF-1 targeted cancer therapeutics.


Clinical & Experimental Metastasis | 1999

Differential modulation of proliferation, matrix metalloproteinase expression and invasion of human head and neck squamous carcinoma cells by c-erbB ligands.

Pornchai O-charoenrat; Peter Rhys-Evans; William Court; Gary Box; Suzanne A. Eccles

Evidence suggests that there is an association between the abnormal expression of members of the c-erbB receptor tyrosine kinase family and poor prognosis in head and neck squamous cell carcinomas (HNSCC). Until now, the relative contributions of different c-erbB ligands to HNSCC progression have not been clearly defined. In this paper we examined the effects of ligands with different c-erbB receptor specificities in terms of their stimulation of HNSCC proliferation, expression of matrix metalloproteinases (MMPs) and invasion. Heregulin-beta1 (HRG-β1; selective c-erbB3/B4 ligand) was found to stimulate proliferation in the majority of cell lines, whereas epidermal growth factor (EGF; EGFR ligand) and betacellulin (BTC; EGFR/B4 ligand) induced variable responses. All three ligands up-regulated multiple MMPs including collagenases, stromelysins, matrilysin and gelatinase B (MMP-9) but had minimal or no effects on gelatinase A (MMP-2), MT1-MMP and tissue inhibitors of MMPs (TIMPs). MMP-9 mRNA was induced to a higher level than other MMPs, although with slower kinetics. HRG-β1 was less active than EGF and BTC at the optimal concentration (relative potency of EGF:BTC:HRG = 3:4:1). In vitro invasion through Matrigel was also increased by all three ligands in proportion to their MMP up-regulation. A specific anti-EGFR monoclonal antibody (mAb ICR62) inhibited MMP up-regulation, migration and invasion induced by all three ligands, whereas an anti-c-erbB-2 mAb ICR12 inhibited mitogenic and motogenic responses following ligand stimulation but had no effect on MMP expression. These results suggest that c-erbB ligands may differentially potentiate the invasive phenotype of HNSCC via co-operative induction of cell proliferation, migration and proteolysis. The EGFR signalling pathway appears to be the dominant component controlling the proteolytic and invasive phenotype in HNSCC, whereas the c-erbB-2 signalling pathway is responsible, in part, for the mitogenic and motogenic effects of ligands.


Cancer Research | 2007

Syk Tyrosine Kinase Is Linked to Cell Motility and Progression in Squamous Cell Carcinomas of the Head and Neck

Sutima Luangdilok; Carol Box; Lisa Patterson; William Court; Kevin J. Harrington; Lisa Pitkin; Peter Rhŷs-Evans; Pornchai O-charoenrat; Suzanne A. Eccles

Syk, a non-receptor tyrosine kinase, is an important component of immunoreceptor signaling in hematopoietic cells. It has been implicated in key regulatory pathways including phosphoinositide 3-kinase and phospholipase Cgamma (PLCgamma) activation in B cells and integrin signaling in platelets and bronchial epithelial cells. Recently, potential roles in cancer have been reported. In breast cancers, reduced Syk expression was associated with invasion, and its overexpression in cell lines was shown to inhibit cell motility. In contrast, Syk has been shown to mediate chemomigration in nasopharyngeal carcinoma cells. Its role in squamous cell carcinomas of the head and neck (SCCHN) has not yet been investigated. Syk mRNA and protein expression was detected in 6 of 10 SCCHN cell lines. When Syk was transfected into Syk-negative cells (SIHN-011A), chemomigration was enhanced in vitro and this was associated with activation of PLCgamma1. Conversely, abrogation of Syk activity by pharmacologic inhibition or small interfering RNA in HN6 cells with high levels of endogenous expression inhibited migration, haptotaxis, and engagement with matrix proteins; this was accompanied by decreased levels of phosphorylated AKT. Similar effects were seen in Syk-positive CAL 27 cells but not in Syk-negative SIHN-011A cells. Immunoprecipitation suggested co-association of Syk with epidermal growth factor receptor and GRB-2. Syk expression in SCCHN patient tissues was examined by semiquantitative real-time PCR (n = 45) and immunohistochemistry (n = 38) in two independent cohorts. Higher levels of Syk expression were observed in tumors and lymph node metastases relative to normal tissues. High Syk expression significantly correlated with worse survival and may be of prognostic value in SCCHN due to its potential role in cell migration and invasion.


American Journal of Pathology | 2009

Brk Protects Breast Cancer Cells from Autophagic Cell Death Induced by Loss of Anchorage

Amanda Harvey; Caroline J. Pennington; Sarah Porter; Rajpal S. Burmi; Dylan R. Edwards; William Court; Suzanne A. Eccles; Mark R. Crompton

Brk, a tyrosine kinase expressed in a majority of breast tumors, but not normal mammary tissue, promotes breast carcinoma cell proliferation. Normal epithelial cells are dependent on cell-cell or cell-matrix interactions for survival and undergo apoptosis after disruption of these interactions. Tumor cells are less sensitive to the induction of apoptosis and are predicted to have the potential to disseminate. We investigated whether Brk has further roles in breast tumor progression by relating its expression to tumor grade and demonstrating its role in the regulation of carcinoma cell survival under non-adherent conditions. Brk expression was determined by reverse transcription PCR on RNA extracted from surgical samples of human breast cancers. Breast carcinoma cell survival in suspension culture was examined when Brk protein levels were suppressed by RNA interference. Additionally, the effect of experimentally overexpressing Brk in otherwise Brk-negative breast carcinoma cells was assessed. Brk mRNA expression was notably higher in grade 3 breast tumors, as compared with lower tumor grades. In suspension culture, Brk suppression increased the rate of cell death, as compared with controls, and this cell death program exhibited characteristics of autophagy but not of apoptosis. Conversely, experimental expression of Brk in Brk-negative cells increased cell survival whereas kinase-inactive Brk did not. Therefore, Brk enhances breast carcinoma cell survival in suspension, suggesting a role for Brk in supporting breast cancer cell dissemination.


Journal of Medicinal Chemistry | 2015

Discovery of Potent, Orally Bioavailable, Small-Molecule Inhibitors of WNT Signaling from a Cell-Based Pathway Screen

Aurélie Mallinger; Simon Crumpler; Mark Pichowicz; Dennis Waalboer; Mark Stubbs; Olajumoke Adeniji-Popoola; Bozena Wood; Elizabeth L. Smith; Ching Thai; Alan T. Henley; Katrin Georgi; William Court; Steve Hobbs; Gary Box; Maria-Jesus Ortiz-Ruiz; Melanie Valenti; Alexis de Haven Brandon; Robert TePoele; Birgitta Leuthner; Paul Workman; Wynne Aherne; Oliver Poeschke; Trevor Clive Dale; Dirk Wienke; Christina Esdar; Felix Rohdich; Florence I. Raynaud; Paul A. Clarke; Suzanne A. Eccles; Frank Stieber

WNT signaling is frequently deregulated in malignancy, particularly in colon cancer, and plays a key role in the generation and maintenance of cancer stem cells. We report the discovery and optimization of a 3,4,5-trisubstituted pyridine 9 using a high-throughput cell-based reporter assay of WNT pathway activity. We demonstrate a twisted conformation about the pyridine–piperidine bond of 9 by small-molecule X-ray crystallography. Medicinal chemistry optimization to maintain this twisted conformation, cognisant of physicochemical properties likely to maintain good cell permeability, led to 74 (CCT251545), a potent small-molecule inhibitor of WNT signaling with good oral pharmacokinetics. We demonstrate inhibition of WNT pathway activity in a solid human tumor xenograft model with evidence for tumor growth inhibition following oral dosing. This work provides a successful example of hypothesis-driven medicinal chemistry optimization from a singleton hit against a cell-based pathway assay without knowledge of the biochemical target.


Cell Biochemistry and Biophysics | 1994

Preclinical models for the evaluation of targeted therapies of metastatic disease.

Suzanne A. Eccles; Gary Box; William Court; Sandle J; Christopher J. Dean

It has been estimated that approx 60–70% of cancer patients harbor overt or subclinical metastases at diagnosis, and it is the eradication of such systemic disease that largely determines survival. Preclinical tumor model systems employed to evaluate potential new treatment strategies should aim to represent the process and patterns of metastasis of their clinical counterparts as closely as possible. Severe combined immune-deficient (SCID) andnu/nu mice have been extensively used as hosts for the growth of human tumor cell lines and in some cases fresh tumor material. However, in most instances the resulting neoplasms fail to metastasize, and the aberrant immune systems of such animals has limited their use mainly to passive therapies of localized disease. Recently, the development of specially selected tumor variants and the use of appropriate orthotopic sites for implantation has provided several models in which dissemination can be demonstrated. Where the gene coding for a potential target antigen has been cloned, and where its overexpression or mutation is associated with malignancy (e.g., c-erbB-2, H-ras), transgenic mice may yield tumors that will develop in these immunocompetent hosts. In some cases such tumors exhibit metastasis. A third approach is to transfect human genes of interest into appropriate rodent tumors expressing the desired metastatic phenotype. These various approaches are compared with particular reference to mammary carcinoma biology.


Bioorganic & Medicinal Chemistry | 2009

The identification of novel PLC-gamma inhibitors using virtual high throughput screening.

Jóhannes Reynisson; William Court; Ciaran O'neill; James E. H. Day; Lisa Patterson; Edward McDonald; Paul Workman; Matilda Katan; Suzanne A. Eccles

Phospholipase C-gamma (PLC-gamma) has been identified as a possible biological target for anticancer drug therapy but suitable inhibitors are lacking. Therefore, in order to identify active compounds (hits) virtual high throughput screening was performed. The crystal structure of the PLC-delta isoform was used as a model docking scaffold since no crystallographic data are available on its gamma counterpart. A pilot screen was performed using approximately 9.2x10(4) compounds, where the robustness of the methodology was tested. This was followed by the main screening effort where approximately 4.4x10(5) compounds were used. In both cases, plausible compounds were identified (virtual hits) and a selection of these was experimentally tested. The most potent compounds were in the single digit micro-molar range as determined from the biochemical (Flashplate) assay. This translated into approximately 15 microM in a functional assay in cells. About 30% of the virtual hits showed activity against PLC-gamma (IC(50)<50 microM).

Collaboration


Dive into the William Court's collaboration.

Top Co-Authors

Avatar

Suzanne A. Eccles

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Gary Box

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Lisa Patterson

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul Workman

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Dean

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Cara Lomas

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Melanie Valenti

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Peter Rhys-Evans

The Royal Marsden NHS Foundation Trust

View shared research outputs
Researchain Logo
Decentralizing Knowledge