Network


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

Hotspot


Dive into the research topics where Emmy D.G. Fleuren is active.

Publication


Featured researches published by Emmy D.G. Fleuren.


The EMBO Journal | 2008

Phosphorylation‐dependent binding of 14‐3‐3 terminates signalling by the Gab2 docking protein

Tilman Brummer; Mark Larance; Maria Teresa Herrera Abreu; Ruth J. Lyons; Paul Timpson; Christoph H Emmerich; Emmy D.G. Fleuren; Gillian M. Lehrbach; Daniel Schramek; Michael Guilhaus; David E. James; Roger J. Daly

Grb2‐associated binder (Gab)2 functions downstream of a variety of receptor and cytoplasmic tyrosine kinases as a docking platform for specific signal transducers and performs important functions in both normal physiology and oncogenesis. Gab2 signalling is promoted by its association with specific receptors through the adaptor Grb2. However, the molecular mechanisms that attenuate Gab2 signals have remained unclear. We now demonstrate that growth factor‐induced phosphorylation of Gab2 on two residues, S210 and T391, leads to recruitment of 14‐3‐3 proteins. Together, these events mediate negative‐feedback regulation, as Gab2S210A/T391A exhibits sustained receptor association and signalling and promotes cell proliferation and transformation. Importantly, introduction of constitutive 14‐3‐3‐binding sites into Gab2 renders it refractory to receptor activation, demonstrating that site‐selective binding of 14‐3‐3 proteins is sufficient to terminate Gab2 signalling. Furthermore, this is associated with reduced binding of Grb2. This leads to a model where signal attenuation occurs because 14‐3‐3 promotes dissociation of Gab2 from Grb2, and thereby uncouples Gab2 from the receptor complex. This represents a novel regulatory mechanism with implications for diverse tyrosine kinase signalling systems.


Clinical Cancer Research | 2011

Predicting IGF-1R therapy response in bone sarcomas: immuno-SPECT imaging with radiolabeled R1507

Emmy D.G. Fleuren; Yvonne M.H. Versleijen-Jonkers; Addy Cm van de Luijtgaarden; Janneke D.M. Molkenboer-Kuenen; Sandra Heskamp; Melissa H.S. Roeffen; Hanneke W. M. van Laarhoven; Peter J. Houghton; Wim J.G. Oyen; Otto C. Boerman; Winette T. A. van der Graaf

Purpose: To investigate whether indium-111–labeled R1507 (111In-R1507) immuno-SPECT (single—photon emission computed tomography), a novel noninvasive, in vivo screening method to visualize membranous insulin-like growth factor 1 receptor (IGF-1R) expression and accessibility, can be used to predict IGF-1R treatment (R1507) response in bone sarcomas. Experimental Design: BALB/c nude mice were subcutaneously implanted with IGF-1R–expressing human bone sarcoma xenografts (OS-1, EW-5, and EW-8) which showed high, modest, or no response, respectively, to R1507, a monoclonal antibody targeting the extracellular domain of IGF-1R. An IGF-1R–negative tumor (OS-33), unresponsive to IGF-1R inhibitors, was examined as well. Mice were injected with 111In-R1507. Biodistribution and immuno-SPECT/computed tomography imaging studies were carried out 1, 3, and 7 days p.i. in mice with OS-1 and EW-5 xenografts and 3 days p.i. in mice with EW-8 and OS-33 xenografts. Results: Biodistribution studies showed specific accumulation of 111In-R1507 in OS-1 and EW-5 xenografts (27.5 ± 6.5%ID/g and 14.0 ± 2.8%ID/g, 3 days p.i., respectively). Most importantly, 111In-R1507 uptake in IGF-1R positive, but unresponsive, EW-8 xenografts (6.5 ± 1.5%ID/g, 3 days p.i.) was similar to that of the IGF-1R–negative OS-33 tumor (5.5 ± 0.6%ID/g, 3 days p.i.). Uptake in normal tissues was low and nonspecific. Corresponding immuno-SPECT images clearly discriminated between high, modest, and nonresponding tumors by showing a homogeneous (OS-1), heterogeneous (EW-5), or nonspecific (EW-8 and OS-33) tumor uptake of 111In-R1507. Conclusions: 111In-R1507 immuno-SPECT is an excellent method to visualize membranous IGF-1R expression and target accessibility in vivo in human bone sarcoma xenografts and may serve as an independent marker to predict IGF-1R therapy (R1507) response in bone sarcoma patients. Clin Cancer Res; 17(24); 7693–703. ©2011 AACR.


International Journal of Cancer | 2010

Overexpression of the oncogenic signal transducer Gab2 occurs early in breast cancer development

Emmy D.G. Fleuren; Sandra A O'Toole; Ewan K.A. Millar; Catriona M. McNeil; Elena Lopez-Knowles; Alice Boulghourjian; David R. Croucher; Daniel Schramek; Tilman Brummer; Josef M. Penninger; Robert L. Sutherland; Roger J. Daly

Gab2, a docking‐type signaling protein with demonstrated oncogenic potential, is overexpressed in breast cancer, but its prognostic significance and role in disease evolution remain unclear. Immunohistochemical detection of Gab2 in a large cohort of primary human breast cancers of known outcome revealed that while Gab2 expression was positively correlated with increased tumor grade, it did not correlate with disease recurrence or breast cancer‐related death in the total cohort or in patients stratified according to lymph node, estrogen receptor (ER) or HER2 status. Interestingly, analysis of a “progression series” that included premalignant and preinvasive breast lesions as well as samples of metastatic disease revealed that Gab2 expression was significantly enhanced in the earliest lesion examined, usual ductal hyperplasia, with a further increase detected in ductal carcinoma in situ (DCIS). Furthermore, expression was less in invasive cancers and lymph node metastases than in DCIS, but still higher than in normal breast. These findings indicate that while Gab2 expression is not prognostic in breast cancer, its role in early disease evolution warrants further analysis, as Gab2 and its effectors may provide targets for novel strategies aimed at preventing breast cancer development.


Molecular Oncology | 2014

Theranostic applications of antibodies in oncology

Emmy D.G. Fleuren; Yvonne M.H. Versleijen-Jonkers; Sandra Heskamp; Carla M.L. van Herpen; Wim J.G. Oyen; Winette T. A. van der Graaf; Otto C. Boerman

Targeted therapies, including antibodies, are becoming increasingly important in cancer therapy. Important limitations, however, are that not every patient benefits from a specific antibody therapy and that responses could be short‐lived due to acquired resistance. In addition, targeted therapies are quite expensive and are not completely devoid of side‐effects. This urges the need for accurate patient selection and response monitoring.


International Journal of Cancer | 2013

Expression and clinical relevance of MET and ALK in Ewing sarcomas

Emmy D.G. Fleuren; Melissa H.S. Roeffen; William Leenders; Uta Flucke; Myrella Vlenterie; Hendrik W. Schreuder; Otto C. Boerman; Winette T. A. van der Graaf; Yvonne M.H. Versleijen-Jonkers

Because novel therapeutic options are limited in Ewing sarcomas (ES), we investigated the expression, genetic aberrations and clinical relevance of MET and anaplastic lymphoma kinase (ALK) in ES and determined the relevance of targeting these receptors. MET and ALK protein expression was determined immunohistochemically in 31 (50 samples) and 36 (59 samples) ES patients, respectively. Samples included primary tumors, postchemotherapy resections, metastases and relapses. MET and ALK RTK domains were sequenced in respectively 33 and 32 tumors. Five ES cell lines were treated in vitro with the MET/ALK‐inhibitor crizotinib, the ALK‐inhibitor NVP‐TAE684 or the MET‐inhibitor cabozantinib and analyzed by MTT assays. Modest to high MET and ALK expression was detected in the majority of ES (86 and 69%, respectively). ALK expression was significantly lower in postchemotherapy resections compared to paired untreated primary tumors (p = 0.031, z = −2.310, n = 11). In primary tumors (n = 20), membranous MET expression significantly correlated with a poor overall survival (OS) (60 vs. 197 months, p = 0.014). There was a trend toward a poor event‐free survival (67 vs. 111 months, p = 0.078) and OS (88 vs. 128 months, p = 0.074) in patients with highest ALK levels (n = 29). ALK or MET RTK domain aberrations were demonstrated in 5/32 (16%) and 3/33 (9%) tumors, respectively. Crizotinib (IC50 1.22–3.59 μmol/L), NVP‐TAE684 (IC50 0.15–0.79 μmol/L) and cabozantinib (IC50 2.69–8.27 μmol/L) affected ES cell viability in vitro. Altogether, our data suggest that MET and ALK are potential novel therapeutic targets in ES and targeting these receptors may be of great interest to rationally design future studies in ES.


Biochimica et Biophysica Acta | 2014

Targeting receptor tyrosine kinases in osteosarcoma and Ewing sarcoma: current hurdles and future perspectives.

Emmy D.G. Fleuren; Yvonne M.H. Versleijen-Jonkers; Otto C. Boerman; Winette T. A. van der Graaf

Osteosarcoma (OS) and Ewing sarcoma (ES) are the two most common types of primary bone cancer, which mainly affect children and young adults. Despite intensive multi-modal treatment, the survival of both OS and ES has not improved much during the last decades and new therapeutic options are awaited. One promising approach is the specific targeting of transmembrane receptor tyrosine kinases (RTKs) implicated in these types of bone cancer. However, despite encouraging in vitro and in vivo results, apart from intriguing results of Insulin-like Growth Factor-1 Receptor (IGF-1R) antibodies in ES, clinical studies are limited or disappointing. Primary resistance to RTK inhibitors is frequently observed in OS and ES patients, and even patients that initially respond well eventually develop acquired resistance. There are, however, a few remarks to make concerning the current set-up of clinical trials and about strategies to improve RTK-based treatments in OS and ES. This review provides an overview concerning current RTK-mediated therapies in OS and ES and discusses the problems observed in the clinic. More importantly, we describe several strategies to overcome resistance to RTK inhibitors which may significantly improve outcome of OS and ES patients.


International Journal of Cancer | 2014

Temsirolimus combined with cisplatin or bevacizumab is active in osteosarcoma models.

Emmy D.G. Fleuren; Yvonne M.H. Versleijen-Jonkers; Melissa H.S. Roeffen; Gerben M. Franssen; Uta Flucke; Peter J. Houghton; Wim J.G. Oyen; Otto C. Boerman; Winette T. A. van der Graaf

Mammalian target of rapamycin (mTOR) is a new promising oncological target. However, most clinical studies reported only modest antitumor activity during mTOR‐targeted monotherapies, including studies in osteosarcomas, emphasizing a need for improvement. We hypothesized that the combination with rationally selected other therapeutic agents may improve response. In this study, we examined the efficacy of the mTOR inhibitor temsirolimus combined with cisplatin or bevacizumab on the growth of human osteosarcoma xenografts (OS‐33 and OS‐1) in vivo, incorporating functional imaging techniques and microscopic analyses to unravel mechanisms of response. In both OS‐33 and OS‐1 models, the activity of temsirolimus was significantly enhanced by the addition of cisplatin (TC) or bevacizumab (TB). Extensive immunohistochemical analysis demonstrated apparent effects on tumor architecture, vasculature, apoptosis and the mTOR‐pathway with combined treatments. 3′‐Deoxy‐3′‐18F‐fluorothymidine (18F‐FLT) positron emission tomography (PET) scans showed a remarkable decrease in 18F‐FLT signal in TC‐ and TB‐treated OS‐1 tumors, which was already noticeable after 1 week of treatment. No baseline uptake was observed in the OS‐33 model. Both immunohistochemistry and 18F‐FLT‐PET demonstrated that responses as determined by caliper measurements underestimated the actual tumor response. Although 18F‐FLT‐PET could be used for accurate and early response monitoring for temsirolimus‐based therapies in the OS‐1 model, we could not evaluate OS‐33 tumors with this molecular imaging technique. Further research on the value of the use of 18F‐FLT‐PET in this setting in osteosarcomas is warranted. Overall, these findings urge the further exploration of TC and TB treatment for osteosarcoma (and other cancer) patients.


Expert Opinion on Investigational Drugs | 2017

Directing the use of DDR kinase inhibitors in cancer treatment

Inger Brandsma; Emmy D.G. Fleuren; Chris T. Williamson; Christopher J. Lord

ABSTRACT Introduction: Defects in the DNA damage response (DDR) drive the development of cancer by fostering DNA mutation but also provide cancer-specific vulnerabilities that can be exploited therapeutically. The recent approval of three different PARP inhibitors for the treatment of ovarian cancer provides the impetus for further developing targeted inhibitors of many of the kinases involved in the DDR, including inhibitors of ATR, ATM, CHEK1, CHEK2, DNAPK and WEE1. Areas covered: We summarise the current stage of development of these novel DDR kinase inhibitors, and describe which predictive biomarkers might be exploited to direct their clinical use. Expert opinion: Novel DDR inhibitors present promising candidates in cancer treatment and have the potential to elicit synthetic lethal effects. In order to fully exploit their potential and maximize their utility, identifying highly penetrant predictive biomarkers of single agent and combinatorial DDR inhibitor sensitivity are critical. Identifying the optimal drug combination regimens that could used with DDR inhibitors is also a key objective.


Cancer Research | 2017

ATR Is a Therapeutic Target in Synovial Sarcoma

Samuel E. Jones; Emmy D.G. Fleuren; Jessica Frankum; Asha Konde; Chris T. Williamson; Dragomir B. Krastev; Helen N. Pemberton; James Campbell; Aditi Gulati; Richard Elliott; Malini Menon; Joanna Selfe; Rachel Brough; Stephen J. Pettitt; Wojciech Niedzwiedz; Winette T. A. van der Graaf; Janet Shipley; Alan Ashworth; Christopher J. Lord

Synovial sarcoma (SS) is an aggressive soft-tissue malignancy characterized by expression of SS18-SSX fusions, where treatment options are limited. To identify therapeutically actionable genetic dependencies in SS, we performed a series of parallel, high-throughput small interfering RNA (siRNA) screens and compared genetic dependencies in SS tumor cells with those in >130 non-SS tumor cell lines. This approach revealed a reliance of SS tumor cells upon the DNA damage response serine/threonine protein kinase ATR. Clinical ATR inhibitors (ATRi) elicited a synthetic lethal effect in SS tumor cells and impaired growth of SS patient-derived xenografts. Oncogenic SS18-SSX family fusion genes are known to alter the composition of the BAF chromatin-remodeling complex, causing ejection and degradation of wild-type SS18 and the tumor suppressor SMARCB1. Expression of oncogenic SS18-SSX fusion proteins caused profound ATRi sensitivity and a reduction in SS18 and SMARCB1 protein levels, but an SSX18-SSX1 Δ71-78 fusion containing a C-terminal deletion did not. ATRi sensitivity in SS was characterized by an increase in biomarkers of replication fork stress (increased γH2AX, decreased replication fork speed, and increased R-loops), an apoptotic response, and a dependence upon cyclin E expression. Combinations of cisplatin or PARP inhibitors enhanced the antitumor cell effect of ATRi, suggesting that either single-agent ATRi or combination therapy involving ATRi might be further assessed as candidate approaches for SS treatment. Cancer Res; 77(24); 7014-26. ©2017 AACR.


Clinical sarcoma research | 2016

The ENCCA-WP7/EuroSarc/EEC/PROVABES/EURAMOS 3rd European Bone Sarcoma Networking Meeting/Joint Workshop of EU Bone Sarcoma Translational Research Networks; Vienna, Austria, September 24-25, 2015. Workshop Report

Leo Kager; Jeremy Whelan; Uta Dirksen; Bass Hassan; Jakob K. Anninga; Lindsey Bennister; Judith V. M. G. Bovée; Bernadette Brennan; Javier Martin Broto; Laurence Brugières; Anne-Marie Cleton-Jansen; Christopher Copland; Aurélie Dutour; Franca Fagioli; Stefano Ferrari; Marta Fiocco; Emmy D.G. Fleuren; Nathalie Gaspar; Hans Gelderblom; Craig Gerrand; Joachim Gerß; Ornella Gonzato; Winette T. A. van der Graaf; Stefanie Hecker-Nolting; David Herrero-Martín; Stephanie Klco-Brosius; Heinrich Kovar; Ruth Ladenstein; Carlo Lancia; Marie-cecile Ledeley

This report summarizes the results of the 3rd Joint ENCCA-WP7, EuroSarc, EEC, PROVABES, and EURAMOS European Bone Sarcoma Network Meeting, which was held at the Children’s Cancer Research Institute in Vienna, Austria on September 24–25, 2015. The joint bone sarcoma network meetings bring together European bone sarcoma researchers to present and discuss current knowledge on bone sarcoma biology, genetics, immunology, as well as results from preclinical investigations and clinical trials, to generate novel hypotheses for collaborative biological and clinical investigations. The ultimate goal is to further improve therapy and outcome in patients with bone sarcomas.

Collaboration


Dive into the Emmy D.G. Fleuren's collaboration.

Top Co-Authors

Avatar

Winette T. A. van der Graaf

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Yvonne M.H. Versleijen-Jonkers

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Otto C. Boerman

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Melissa H.S. Roeffen

Radboud University Nijmegen Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Lord

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Uta Flucke

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Chris T. Williamson

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Wim J.G. Oyen

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Janet Shipley

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jessica Frankum

Institute of Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge