Network


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

Hotspot


Dive into the research topics where Nicola E. A. Chessum is active.

Publication


Featured researches published by Nicola E. A. Chessum.


Progress in Medicinal Chemistry | 2015

Recent advances in cancer therapeutics.

Nicola E. A. Chessum; Keith Jones; Elisa Pasqua; Michael Tucker

In the past 20 years, cancer therapeutics has undergone a paradigm shift away from the traditional cytotoxic drugs towards the targeting of proteins intimately involved in driving the cancer phenotype. The poster child for this alternative approach to the treatment of cancer is imatinib, a small-molecule kinase inhibitor designed to target chronic myeloid leukaemia driven by the BCR-ABL translocation in a defined patient population. The improvement in survival achieved by treatment of this patient cohort with imatinib is impressive. Thus, the aim is to provide efficacy but with low toxicity. The role of the medicinal chemist in oncology drug discovery is now closely aligned with the role in most other therapeutic areas with high-throughput and/or fragment-based screening, structure-based design, selectivity, pharmacokinetic optimisation and pharmacodynamic biomarker modulation, all playing a familiar part in the process. In this chapter, we selected four areas in which compounds are either approved drugs or in clinical trials. These are chaperone inhibitors, kinase inhibitors, histone deacetylase inhibitors and inhibitors of protein-protein interactions. Even within these areas, we have been selective, particularly for kinase inhibitors, and our aim has been to exemplify newer approaches and novel aspects of medicinal chemistry.


Journal of Medicinal Chemistry | 2017

Discovery of a Chemical Probe Bisamide (CCT251236): An Orally Bioavailable Efficacious Pirin Ligand from a Heat Shock Transcription Factor 1 (HSF1) Phenotypic Screen

Matthew D. Cheeseman; Nicola E. A. Chessum; Carl S. Rye; A. Elisa Pasqua; Michael Tucker; Birgit Wilding; Lindsay E. Evans; Susan Lepri; Meirion Richards; Swee Y. Sharp; Salyha Ali; Martin G. Rowlands; Lisa O’Fee; Asadh Miah; Angela Hayes; Alan T. Henley; Marissa V. Powers; Robert te Poele; Emmanuel de Billy; Loredana Pellegrino; Florence I. Raynaud; Rosemary Burke; Rob L. M. van Montfort; Suzanne A. Eccles; Paul Workman; Keith Jones

Phenotypic screens, which focus on measuring and quantifying discrete cellular changes rather than affinity for individual recombinant proteins, have recently attracted renewed interest as an efficient strategy for drug discovery. In this article, we describe the discovery of a new chemical probe, bisamide (CCT251236), identified using an unbiased phenotypic screen to detect inhibitors of the HSF1 stress pathway. The chemical probe is orally bioavailable and displays efficacy in a human ovarian carcinoma xenograft model. By developing cell-based SAR and using chemical proteomics, we identified pirin as a high affinity molecular target, which was confirmed by SPR and crystallography.


Clinical Cancer Research | 2018

HSF1 Is Essential for Myeloma Cell Survival and A Promising Therapeutic Target

Jacqueline H.L. Fok; Somaieh Hedayat; Lei Zhang; Lauren I. Aronson; Fabio Mirabella; Charlotte Pawlyn; Michael D. Bright; Christopher P. Wardell; Jonathan J. Keats; Emmanuel de Billy; Carl S. Rye; Nicola E. A. Chessum; Keith Jones; Gareth J. Morgan; Suzanne A. Eccles; Paul Workman; Faith E. Davies

Purpose: Myeloma is a plasma cell malignancy characterized by the overproduction of immunoglobulin, and is therefore susceptible to therapies targeting protein homeostasis. We hypothesized that heat shock factor 1 (HSF1) was an attractive therapeutic target for myeloma due to its direct regulation of transcriptional programs implicated in both protein homeostasis and the oncogenic phenotype. Here, we interrogate HSF1 as a therapeutic target in myeloma using bioinformatic, genetic, and pharmacologic means. Experimental Design: To assess the clinical relevance of HSF1, we analyzed publicly available patient myeloma gene expression datasets. Validation of this novel target was conducted in in vitro experiments using shRNA or inhibitors of the HSF1 pathway in human myeloma cell lines and primary cells as well as in in vivo human myeloma xenograft models. Results: Expression of HSF1 and its target genes were associated with poorer myeloma patient survival. ShRNA-mediated knockdown or pharmacologic inhibition of the HSF1 pathway with a novel chemical probe, CCT251236, or with KRIBB11, led to caspase-mediated cell death that was associated with an increase in EIF2α phosphorylation, CHOP expression and a decrease in overall protein synthesis. Importantly, both CCT251236 and KRIBB11 induced cytotoxicity in human myeloma cell lines and patient-derived primary myeloma cells with a therapeutic window over normal cells. Pharmacologic inhibition induced tumor growth inhibition and was well-tolerated in a human myeloma xenograft murine model with evidence of pharmacodynamic biomarker modulation. Conclusions: Taken together, our studies demonstrate the dependence of myeloma cells on HSF1 for survival and support the clinical evaluation of pharmacologic inhibitors of the HSF1 pathway in myeloma. Clin Cancer Res; 24(10); 2395–407. ©2018 AACR. See related commentary by Parekh, p. 2237


Journal of Medicinal Chemistry | 2018

Demonstrating In-Cell Target Engagement Using a Pirin Protein Degradation Probe (CCT367766)

Nicola E. A. Chessum; Swee Y. Sharp; John Caldwell; A. Elisa Pasqua; Birgit Wilding; Giampiero Colombano; Ian Collins; Bugra Ozer; Meirion Richards; Martin G. Rowlands; Mark Stubbs; Rosemary Burke; P. Craig McAndrew; Paul A. Clarke; Paul Workman; Matthew D. Cheeseman; Keith Jones

Demonstrating intracellular protein target engagement is an essential step in the development and progression of new chemical probes and potential small molecule therapeutics. However, this can be particularly challenging for poorly studied and noncatalytic proteins, as robust proximal biomarkers are rarely known. To confirm that our recently discovered chemical probe 1 (CCT251236) binds the putative transcription factor regulator pirin in living cells, we developed a heterobifunctional protein degradation probe. Focusing on linker design and physicochemical properties, we generated a highly active probe 16 (CCT367766) in only three iterations, validating our efficient strategy for degradation probe design against nonvalidated protein targets.


Cancer Research | 2017

Abstract LB-304: Discovery of chemical probe CCT251236: An orally bioavailable efficacious pirin ligand from an HSF1 phenotypic screen

Matthew D. Cheeseman; Nicola E. A. Chessum; Carl S. Rye; Elisa Pasqua; Michael Tucker; Birgit Wilding; Lindsay E. Evans; Susan Lepri; Meirion Richards; Swee Y. Sharp; Salyha Ali; Martin G. Rowlands; Lisa O'Fee; Asadh Miah; Angela Hayes; Alan T. Henley; Marissa V. Powers; Robert te Poele; Emmanuel de Billy; Loredana Pellegrino; Florence I. Raynaud; Rosemary Burke; Rob L. M. van Montfort; Suzanne A. Eccles; Keith Jones; Paul Workman

Heat shock factor 1 (HSF1) was originally identified as a master regulator of the classical ‘cytoprotective’ heat shock response. However, a large body of evidence has now verified the importance of HSF1 to tumorigenesis and cancer progression. HSF1 is activated by various elements of the cancer state, reprogramming the transcriptome in a way that is overlapping with, but distinct from, the canonical heat-shock response. Also, there is a strong correlation between the expression of activated HSF1 in tumors and adverse clinical outcomes. This evidence indicates that the inhibition of HSF1-mediated transcription could be a viable strategy in cancer treatment. Inhibiting the HSF1 stress pathway represents an attempt at targeting non-oncogene addiction and proteotoxic stress, which has been proposed to be advantageous. However, HSF1 is a ligandless transcription factor and is unlikely to be amenable to standard drug discovery strategies and direct inhibition with small molecules. Therefore, we proposed that inhibitors of HSF1-mediated transcription, which antagonize the HSF1 pathway but without necessarily binding directly to HSF1, could be discovered and developed via a cell-based phenotypic screen. We carried out a high throughput Arrayscan assay of 200,000 compounds to measure the inhibition of HSF1-mediated HSP72 expression stimulated by pre-treatment with an HSP90 inhibitor. We identified a singleton hit with a bisamide core, CCT245232. This compound showed potent growth inhibition in a range of human cancer cell lines but had poor physicochemical properties leading to an unacceptable pharmacokinetic profile. Improvement of the physicochemical properties of CCT245232 whilst maintaining potency versus our cell-based assays led to the orally bioavailable tool compound CCT251236. This compound shows potent growth inhibition (GI50 values in low nanomolar range) of human ovarian cancer cell lines in vitro and good efficacy against human ovarian cancer xenografts in nude mice in vivo. We applied chemo-proteomic strategies to identify the molecular target using a probe based on CCT251236 and discovered pirin as a high affinity molecular target. Binding of CCT251236 to recombinant pirin was confirmed in biophysical assays. CCT251236 recapitulates the reported anti-migratory phenotype for a pirin ligand although binding to pirin alone does not explain the cellular phenotype observed with our chemical tool. We are currently using CCT251236 as a chemical probe while further optimizing its properties to identify a clinical candidate. Citation Format: Matthew D. Cheeseman, Nicola E. Chessum, Carl S. Rye, Elisa A. Pasqua, Michael J. Tucker, Birgit Wilding, Lindsay E. Evans, Susan Lepri, Meirion Richards, Swee Y. Sharp, Salyha Ali, Martin Rowlands, Lisa O9Fee, Asadh Miah, Angela Hayes, Alan T. Henley, Marissa Powers, Robert te Poele, Emmanuel De Billy, Loredana Pellegrino, Florence Raynaud, Rosemary Burke, Robert L. van Montfort, Suzanne A. Eccles, Keith Jones, Paul Workman. Discovery of chemical probe CCT251236: An orally bioavailable efficacious pirin ligand from an HSF1 phenotypic screen [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-304. doi:10.1158/1538-7445.AM2017-LB-304


MedChemComm | 2016

Correction: Discovery of 4,6-disubstituted pyrimidines as potent inhibitors of the heat shock factor 1 (HSF1) stress pathway and CDK9. See DOI: 10.1039/c6md00159a

Carl S. Rye; Nicola E. A. Chessum; Scott Lamont; Kurt Gordon Pike; Paul Faulder; Julie Demeritt; Paul D. Kemmitt; Julie A. Tucker; Lorenzo Zani; Matthew D. Cheeseman; Rosie Isaac; Louise Goodwin; Joanna Boros; Florence I. Raynaud; Angela Hayes; Alan T. Henley; Emmanuel de Billy; Christopher J. Lynch; Swee Y. Sharp; Robert te Poele; Lisa O’Fee; Kevin Michael Foote; Stephen Green; Paul Workman; Keith Jones

Correction for ‘Discovery of 4,6-disubstituted pyrimidines as potent inhibitors of the heat shock factor 1 (HSF1) stress pathway and CDK9’ by Carl S. Rye et al., Med. Chem. Commun., 2016, 7, 1580–1586.


Cancer Research | 2014

Abstract 1775: Identification of small molecule inhibitors of HSF1 stress pathway activation in cancer cells

Emmanuel de Billy; Nicola E. A. Chessum; Robert te Poele; Jennifer R. Smith; Lorenzo Zani; Swee Y. Sharp; Mark Stubbs; Wynne Aherne; Keith Jones; Paul Workman

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Heat Shock Factor 1 (HSF1) is a key transcription factor involved in proteostasis and response to stress, as well as being implicated in many diseases including cancer. Up-regulation of its activity by environmental stress or oncogenesis leads to transcriptional induction of genes involved in diverse cellular processes supporting the cancer state, including proteostasis, proliferation, survival and metastasis. Inhibition of HSF1 is therefore potentially beneficial for cancer treatment, but HSF1 is not technically druggable. We developed a high-throughput cell-based reporter gene assay to screen a library of small-molecule kinase inhibitors and identified 3 compound series as validated inhibitors of HSF1 activation by the HSP90 inhibitor 17-AAG. Further characterization of imidazo[1,2-b]pyridazine compounds showed inhibition of HSF1 target gene expression and the ability to mimic other features of the HSF1 knockdown phenotype. Using these tool compounds and by siRNA knockdown of HSF1 expression we demonstrate that HSF1 inhibition leads to cell cycle arrest and enhances the antiproliferative effect of 17-AAG. In addition, we show that HSP90 inhibition induces HSF1 phosphorylation at two serine residues, Ser326 and Ser320, highlighting the importance of Ser320 phosphorylation in HSP72 up-regulation by 17-AAG-induced HSF1 activation, and show that this activation is inhibited by our tool compounds. Our findings support the optimization and development of small-molecule inhibitors of the HSF1 pathway for cancer treatment. Note: This abstract was not presented at the meeting. Citation Format: Emmanuel de Billy, Nicola Chessum, Robert Te Poele, Jennifer Smith, Lorenzo Zani, Swee Sharp, Mark Stubbs, Wynne Aherne, Keith Jones, Paul Workman. Identification of small molecule inhibitors of HSF1 stress pathway activation in cancer cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1775. doi:10.1158/1538-7445.AM2014-1775


Tetrahedron Letters | 2008

An efficient synthesis of 2,3-diaryl (3H)-quinazolin-4-ones via imidoyl chlorides

Andrew Kalusa; Nicola E. A. Chessum; Keith Jones


MedChemComm | 2016

Discovery of 4,6-disubstituted pyrimidines as potent inhibitors of the heat shock factor 1 (HSF1) stress pathway and CDK9

Carl S. Rye; Nicola E. A. Chessum; Scott Lamont; Kurt Gordon Pike; Paul Faulder; Julie Demeritt; Paul D. Kemmitt; Julie A. Tucker; Lorenzo Zani; Matthew D. Cheeseman; Rosie Isaac; Louise Goodwin; Joanna Boros; Florence I. Raynaud; Angela Hayes; Alan T. Henley; Emmanuel de Billy; Christopher J. Lynch; Swee Y. Sharp; Robert te Poele; Lisa O’Fee; Kevin Michael Foote; Stephen Green; Paul Workman; Keith Jones


Organic and Biomolecular Chemistry | 2015

A model β-sheet interaction and thermodynamic analysis of β-strand mimetics

Colin W. Robinson; Carl S. Rye; Nicola E. A. Chessum; Keith Jones

Collaboration


Dive into the Nicola E. A. Chessum's collaboration.

Top Co-Authors

Avatar

Keith Jones

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Matthew D. Cheeseman

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Paul Workman

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Carl S. Rye

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Swee Y. Sharp

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Emmanuel de Billy

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Robert te Poele

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Alan T. Henley

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Angela Hayes

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Birgit Wilding

Institute of Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge