Network


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

Hotspot


Dive into the research topics where Kathryn R. Taylor is active.

Publication


Featured researches published by Kathryn R. Taylor.


Nature Genetics | 2014

Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations

Pawel Buczkowicz; Christine M. Hoeman; Patricia Rakopoulos; Sanja Pajovic; Louis Letourneau; Misko Dzamba; Andrew Morrison; Peter W. Lewis; Eric Bouffet; Ute Bartels; Jennifer Zuccaro; Sameer Agnihotri; Scott Ryall; Mark Barszczyk; Yevgen Chornenkyy; Mathieu Bourgey; Guillaume Bourque; Alexandre Montpetit; Francisco Cordero; Pedro Castelo-Branco; Joshua Mangerel; Uri Tabori; King Ching Ho; Annie Huang; Kathryn R. Taylor; Alan Mackay; Javad Nazarian; Jason Fangusaro; Matthias A. Karajannis; David Zagzag

Diffuse intrinsic pontine glioma (DIPG) is a fatal brain cancer that arises in the brainstem of children, with no effective treatment and near 100% fatality. The failure of most therapies can be attributed to the delicate location of these tumors and to the selection of therapies on the basis of assumptions that DIPGs are molecularly similar to adult disease. Recent studies have unraveled the unique genetic makeup of this brain cancer, with nearly 80% found to harbor a p.Lys27Met histone H3.3 or p.Lys27Met histone H3.1 alteration. However, DIPGs are still thought of as one disease, with limited understanding of the genetic drivers of these tumors. To understand what drives DIPGs, we integrated whole-genome sequencing with methylation, expression and copy number profiling, discovering that DIPGs comprise three molecularly distinct subgroups (H3-K27M, silent and MYCN) and uncovering a new recurrent activating mutation affecting the activin receptor gene ACVR1 in 20% of DIPGs. Mutations in ACVR1 were constitutively activating, leading to SMAD phosphorylation and increased expression of the downstream activin signaling targets ID1 and ID2. Our results highlight distinct molecular subgroups and novel therapeutic targets for this incurable pediatric cancer.


Nature Genetics | 2014

Recurrent activating ACVR1 mutations in diffuse intrinsic pontine glioma.

Kathryn R. Taylor; Alan Mackay; Nathalene Truffaux; Yaron S N Butterfield; Olena Morozova; Cathy Philippe; David Castel; Catherine S. Grasso; Maria Vinci; Diana Carvalho; Angel M. Carcaboso; Carmen Torres; Ofelia Cruz; Jaume Mora; Natacha Entz-Werle; Wendy J. Ingram; Michelle Monje; Darren Hargrave; Alex N. Bullock; Stéphanie Puget; Stephen Yip; Chris Jones; Jacques Grill

Diffuse intrinsic pontine gliomas (DIPGs) are highly infiltrative malignant glial neoplasms of the ventral pons that, due to their location within the brain, are unsuitable for surgical resection and consequently have a universally dismal clinical outcome. The median survival time is 9–12 months, with neither chemotherapeutic nor targeted agents showing substantial survival benefit in clinical trials in children with these tumors. We report the identification of recurrent activating mutations in the ACVR1 gene, which encodes a type I activin receptor serine/threonine kinase, in 21% of DIPG samples. Strikingly, these somatic mutations (encoding p.Arg206His, p.Arg258Gly, p.Gly328Glu, p.Gly328Val, p.Gly328Trp and p.Gly356Asp substitutions) have not been reported previously in cancer but are identical to mutations found in the germ line of individuals with the congenital childhood developmental disorder fibrodysplasia ossificans progressiva (FOP) and have been shown to constitutively activate the BMP–TGF-β signaling pathway. These mutations represent new targets for therapeutic intervention in this otherwise incurable disease.


Cancer Discovery | 2013

Histone H3.3 Mutations Drive Pediatric Glioblastoma through Upregulation of MYCN

Lynn Bjerke; Alan Mackay; Meera Nandhabalan; Anna Burford; Alexa Jury; Sergey Popov; Diana Carvalho; Kathryn R. Taylor; Maria Vinci; Ilirjana Bajrami; Imelda M. McGonnell; Christopher J. Lord; Rui M. Reis; Darren Hargrave; Alan Ashworth; Paul Workman; Chris Jones

UNLABELLED Children and young adults with glioblastoma (GBM) have a median survival rate of only 12 to 15 months, and these GBMs are clinically and biologically distinct from histologically similar cancers in older adults. They are defined by highly specific mutations in the gene encoding the histone H3.3 variant H3F3A , occurring either at or close to key residues marked by methylation for regulation of transcription—K27 and G34. Here, we show that the cerebral hemisphere-specific G34 mutation drives a distinct expression signature through differential genomic binding of the K36 trimethylation mark (H3K36me3). The transcriptional program induced recapitulates that of the developing forebrain, and involves numerous markers of stem-cell maintenance, cell-fate decisions, and self-renewal.Critically, H3F3A G34 mutations cause profound upregulation of MYCN , a potent oncogene that is causative of GBMs when expressed in the correct developmental context. This driving aberration is selectively targetable in this patient population through inhibiting kinases responsible for stabilization of the protein. SIGNIFICANCE We provide the mechanistic explanation for how the fi rst histone gene mutation inhuman disease biology acts to deliver MYCN, a potent tumorigenic initiator, into a stem-cell compartment of the developing forebrain, selectively giving rise to incurable cerebral hemispheric GBM. Using synthetic lethal approaches to these mutant tumor cells provides a rational way to develop novel and highly selective treatment strategies


Clinical Cancer Research | 2013

Dual blockade of the PI3K/AKT/mTOR (AZD8055) and RAS/MEK/ERK (AZD6244) pathways synergistically inhibits rhabdomyosarcoma cell growth in vitro and in vivo.

Jane Renshaw; Kathryn R. Taylor; Ryan Bishop; Melanie Valenti; Alexis de Haven Brandon; Sharon Gowan; Suzanne A. Eccles; Ruth Ruddle; Louise Johnson; Florence I. Raynaud; Joanna Selfe; Khin Thway; Torsten Pietsch; Andrew D.J. Pearson; Janet Shipley

Purpose: To provide rationale for using phosphoinositide 3-kinase (PI3K) and/or mitogen-activated protein kinase (MAPK) pathway inhibitors to treat rhabdomyosarcomas, a major cause of pediatric and adolescent cancer deaths. Experimental Design: The prevalence of PI3K/MAPK pathway activation in rhabdomyosarcoma clinical samples was assessed using immunohistochemistry. Compensatory signaling and cross-talk between PI3K/MAPK pathways was determined in rhabdomyosarcoma cell lines following p110α short hairpin RNA–mediated depletion. Pharmacologic inhibition of reprogrammed signaling in stable p110α knockdown lines was used to determine the target-inhibition profile inducing maximal growth inhibition. The in vitro and in vivo efficacy of inhibitors of TORC1/2 (AZD8055), MEK (AZD6244), and P13K/mTOR (NVP-BEZ235) was evaluated alone and in pairwise combinations. Results: PI3K pathway activation was seen in 82.5% rhabdomyosarcomas with coactivated MAPK in 36% and 46% of alveolar and embryonal subtypes, respectively. p110α knockdown in cell lines over the short and long term was associated with compensatory expression of other p110 isoforms, activation of the MAPK pathway, and cross-talk to reactivate the PI3K pathway. Combinations of PI3K pathway and MAP–ERK kinase (MEK) inhibitors synergistically inhibited cell growth in vitro. Treatment of RD cells with AZD8055 plus AZD6244 blocked reciprocal pathway activation, as evidenced by reduced AKT/ERK/S6 phosphorylation. In vivo, the synergistic effect on growth and changes in pharmacodynamic biomarkers was recapitulated using the AZD8055/AZD6244 combination but not NVP-BEZ235/AZD6244. Pharmacokinetic analysis provided evidence of drug–drug interaction with both combinations. Conclusions: Dual PI3K/MAPK pathway activation and compensatory signaling in both rhabdomyosarcoma subtypes predict a lack of clinical efficacy for single agents targeting either pathway, supporting a therapeutic strategy combining a TORC1/2 with a MEK inhibitor. Clin Cancer Res; 19(21); 5940–51. ©2013 AACR.


Journal of Medicinal Chemistry | 2008

Identification of 4-(4-Aminopiperidin-1-Yl)-7H-Pyrrolo[2,3-D]Pyrimidines as Selective Inhibitors of Protein Kinase B Through Fragment Elaboration.

John Caldwell; Thomas G. Davies; Alastair Donald; Tatiana McHardy; Martin G. Rowlands; G. W Aherne; Lisa-Jane K. Hunter; Kathryn R. Taylor; Ruth Ruddle; Florence I. Raynaud; Marinus Leendert Verdonk; Paul Workman; Garrett; Ian Collins

Fragment-based screening identified 7-azaindole as a protein kinase B inhibitor scaffold. Fragment elaboration using iterative crystallography of inhibitor-PKA-PKB chimera complexes efficiently guided improvements in the potency and selectivity of the compounds, resulting in the identification of nanomolar 6-(piperidin-1-yl)purine, 4-(piperidin-1-yl)-7-azaindole, and 4-(piperidin-1-yl)pyrrolo[2,3- d]pyrimidine inhibitors of PKBbeta with antiproliferative activity and showing pathway inhibition in cells. A divergence in the binding mode was seen between 4-aminomethylpiperidine and 4-aminopiperidine containing molecules. Selectivity for PKB vs PKA was observed with 4-aminopiperidine derivatives, and the most PKB-selective inhibitor (30-fold) showed significantly different bound conformations between PKA and PKA-PKB chimera.


Clinical Cancer Research | 2012

Antitumor Activity of Sustained N-Myc Reduction in Rhabdomyosarcomas and Transcriptional Block by Antigene Therapy

Roberto Tonelli; Alan McIntyre; Consuelo Camerin; Zoë S. Walters; Korinne Di Leo; Joanna Selfe; Stefania Purgato; Edoardo Missiaglia; Andrea Tortori; Jane Renshaw; Annalisa Astolfi; Kathryn R. Taylor; Salvatore Serravalle; Ryan Bishop; Cristina Nanni; Linda J. Valentijn; Andrea Faccini; Ivo Leuschner; Serena Formica; Jorge S. Reis-Filho; Valentina Ambrosini; Khin Thway; Monica Franzoni; Brenda Summersgill; Rosangela Marchelli; Patrizia Hrelia; Giorgio Cantelli-Forti; Stefano Fanti; Roberto Corradini; Andrea Pession

Purpose: Rhabdomyosarcomas are a major cause of cancer death in children, described with MYCN amplification and, in the alveolar subtype, transcription driven by the PAX3-FOXO1 fusion protein. Our aim was to determine the prevalence of N-Myc protein expression and the potential therapeutic effects of reducing expression in rhabdomyosarcomas, including use of an antigene strategy that inhibits transcription. Experimental Design: Protein expression was assessed by immunohistochemistry. MYCN expression was reduced in representative cell lines by RNA interference and an antigene peptide nucleic acid (PNA) oligonucleotide conjugated to a nuclear localization signal peptide. Associated gene expression changes, cell viability, and apoptosis were analyzed in vitro. As a paradigm for antigene therapy, the effects of systemic treatment of mice with rhabdomyosarcoma cell line xenografts were determined. Results: High N-Myc levels were significantly associated with genomic amplification, presence of the PAX3/7-FOXO1 fusion genes, and proliferative capacity. Sustained reduction of N-Myc levels in all rhabdomyosarcoma cell lines that express the protein decreased cell proliferation and increased apoptosis. Positive feedback was shown to regulate PAX3-FOXO1 and N-Myc levels in the alveolar subtype that critically decrease PAX3-FOXO1 levels on reducing N-Myc. Pharmacologic systemic administration of the antigene PNA can eliminate alveolar rhabdomyosarcoma xenografts in mice, without relapse or toxicity. Conclusion: N-Myc, with its restricted expression in non-fetal tissues, is a therapeutic target to treat rhabdomyosarcomas, and blocking gene transcription using antigene oligonucleotide strategies has therapeutic potential in the treatment of cancer and other diseases that has not been previously realized in vivo. Clin Cancer Res; 18(3); 796–807. ©2011 AACR.


Cancer Cell | 2017

Integrated Molecular Meta-Analysis of 1,000 Pediatric High-Grade and Diffuse Intrinsic Pontine Glioma

Alan Mackay; Anna Burford; Diana Carvalho; Elisa Izquierdo; Janat Fazal-Salom; Kathryn R. Taylor; Lynn Bjerke; Matthew Clarke; Mara Vinci; Meera Nandhabalan; Sara Temelso; Sergey Popov; Valeria Molinari; Pichai Raman; Angela J. Waanders; Harry J. Han; Saumya Gupta; Lynley V. Marshall; Stergios Zacharoulis; Sucheta Vaidya; Henry Mandeville; Leslie R. Bridges; Andrew J. Martin; Safa Al-Sarraj; Christopher Chandler; Ho Keung Ng; Xingang Li; Kun Mu; Saoussen Trabelsi; Dorra H’mida-Ben Brahim

Summary We collated data from 157 unpublished cases of pediatric high-grade glioma and diffuse intrinsic pontine glioma and 20 publicly available datasets in an integrated analysis of >1,000 cases. We identified co-segregating mutations in histone-mutant subgroups including loss of FBXW7 in H3.3G34R/V, TOP3A rearrangements in H3.3K27M, and BCOR mutations in H3.1K27M. Histone wild-type subgroups are refined by the presence of key oncogenic events or methylation profiles more closely resembling lower-grade tumors. Genomic aberrations increase with age, highlighting the infant population as biologically and clinically distinct. Uncommon pathway dysregulation is seen in small subsets of tumors, further defining the molecular diversity of the disease, opening up avenues for biological study and providing a basis for functionally defined future treatment stratification.


Cancer Cell | 2017

Transcriptional Dependencies in Diffuse Intrinsic Pontine Glioma.

Surya Nagaraja; Nicholas A. Vitanza; Pamelyn Woo; Kathryn R. Taylor; Fang Liu; Lei Zhang; Meng Li; Wei Meng; Anitha Ponnuswami; Wenchao Sun; Jie Ma; Esther Hulleman; Tomek Swigut; Joanna Wysocka; Yujie Tang; Michelle Monje

Diffuse intrinsic pontine glioma (DIPG) is a fatal pediatric cancer with limited therapeutic options. The majority of cases of DIPG exhibit a mutation in histone-3 (H3K27M) that results in oncogenic transcriptional aberrancies. We show here that DIPG is vulnerable to transcriptional disruption using bromodomain inhibition or CDK7 blockade. Targeting oncogenic transcription through either of these methods synergizes with HDAC inhibition, and DIPG cells resistant to HDAC inhibitor therapy retain sensitivity to CDK7 blockade. Identification of super-enhancers in DIPG provides insights toward the cell of origin, highlighting oligodendroglial lineage genes, and reveals unexpected mechanisms mediating tumor viability and invasion, including potassium channel function and EPH receptor signaling. The findings presented demonstrate transcriptional vulnerabilities and elucidate previously unknown mechanisms of DIPG pathobiology.


Cancer Research | 2014

ACVR1 Mutations in DIPG: Lessons Learned from FOP

Kathryn R. Taylor; Maria Vinci; Alex N. Bullock; Chris Jones

Whole-genome sequencing studies have recently identified a quarter of cases of the rare childhood brainstem tumor diffuse intrinsic pontine glioma to harbor somatic mutations in ACVR1. This gene encodes the type I bone morphogenic protein receptor ALK2, with the residues affected identical to those that, when mutated in the germline, give rise to the congenital malformation syndrome fibrodysplasia ossificans progressiva (FOP), resulting in the transformation of soft tissue into bone. This unexpected link points toward the importance of developmental biology processes in tumorigenesis and provides an extensive experience in mechanistic understanding and drug development hard-won by FOP researchers to pediatric neurooncology. Here, we review the literature in both fields and identify potential areas for collaboration and rapid advancement for patients of both diseases.


The Journal of Pathology | 2018

IGF1R signalling in testicular germ cell tumour cells impacts on cell survival and acquired cisplatin resistance

Joanna Selfe; N. C. Goddard; Alan McIntyre; Kathryn R. Taylor; Jane Renshaw; Sergey Popov; Khin Thway; Brenda Summersgill; Robert Huddart; Duncan C. Gilbert; Janet Shipley

Testicular germ cell tumours (TGCTs) are the most frequent malignancy and cause of death from solid tumours in the 20‐ to 40‐year age group. Although most cases show sensitivity to cis‐platinum‐based chemotherapy, this is associated with long‐term toxicities and chemo‐resistance. Roles for receptor tyrosine kinases other than KIT are largely unknown in TGCT. We therefore conducted a phosphoproteomic screen and identified the insulin growth factor receptor‐1 (IGF1R) as both highly expressed and activated in TGCT cell lines representing the nonseminomatous subtype. IGF1R was also frequently expressed in tumour samples from patients with nonseminomas. Functional analysis of cell line models showed that long‐term shRNA‐mediated IGF1R silencing leads to apoptosis and complete ablation of nonseminoma cells with active IGF1R signalling. Cell lines with high levels of IGF1R activity also showed reduced AKT signalling in response to decreased IGF1R expression as well as sensitivity to the small‐molecule IGF1R inhibitor NVP‐AEW541. These results were in contrast to those in the seminoma cell line TCAM2 that lacked IGF1R signalling via AKT and was one of the two cell lines least sensitive to the IGF1R inhibitor. The dependence on IGF1R activity in the majority of nonseminomas parallels the known role of IGF signalling in the proliferation, migration, and survival of primordial germ cells, the putative cell of origin for TGCT. Upregulation of IGF1R expression and signalling was also found to contribute to acquired cisplatin resistance in an in vitro nonseminoma model, providing a rationale for targeting IGF1R in cisplatin‐resistant disease.

Collaboration


Dive into the Kathryn R. Taylor's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Valeria Molinari

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Alan Mackay

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Chris Jones

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Anna Burford

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Diana Carvalho

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Mara Vinci

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Maria Vinci

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge