Network


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

Hotspot


Dive into the research topics where Christopher Wells is active.

Publication


Featured researches published by Christopher Wells.


Proceedings of the National Academy of Sciences of the United States of America | 2013

RVX-208, an inhibitor of BET transcriptional regulators with selectivity for the second bromodomain

Sarah Picaud; Christopher Wells; I. Felletar; Deborah Brotherton; Sarah Martin; P. Savitsky; Beatriz Diez-Dacal; Martin Philpott; C. Bountra; Hannah Lingard; Oleg Fedorov; Susanne Müller; Paul E. Brennan; Stefan Knapp; Panagis Filippakopoulos

Significance Bromo and extraterminal (BET) proteins have diverse roles in regulating tissue-specific transcriptional programs, raising safety concerns for their inhibition and suggesting that targeting of specific isoforms or even specific domains within this subfamily is important. We report the discovery and characterization of RVX-208 as a domain-selective inhibitor of BETs and provide a potential mechanism of action of a clinical compound that was identified based on phenotypic screens. Bromodomains have emerged as attractive candidates for the development of inhibitors targeting gene transcription. Inhibitors of the bromo and extraterminal (BET) family recently showed promising activity in diverse disease models. However, the pleiotropic nature of BET proteins regulating tissue-specific transcription has raised safety concerns and suggested that attempts should be made for domain-specific targeting. Here, we report that RVX-208, a compound currently in phase II clinical trials, is a BET bromodomain inhibitor specific for second bromodomains (BD2s). Cocrystal structures revealed binding modes of RVX-208 and its synthetic precursor, and fluorescent recovery after photobleaching demonstrated that RVX-208 displaces BET proteins from chromatin. However, gene-expression data showed that BD2 inhibition only modestly affects BET-dependent gene transcription. Our data demonstrate the feasibility of specific targeting within the BET family resulting in different transcriptional outcomes and highlight the importance of BD1 in transcriptional regulation.


Nature Chemical Biology | 2014

Dual kinase-bromodomain inhibitors for rationally designed polypharmacology.

Pietro Ciceri; Susanne Müller; Alison O'Mahony; Oleg Fedorov; Panagis Filippakopoulos; Jeremy P. Hunt; Elisabeth Lasater; Gabriel Pallares; Sarah Picaud; Christopher Wells; Sarah Martin; Lisa Wodicka; Neil P. Shah; Daniel Kelly Treiber; Stefan Knapp

Concomitant inhibition of multiple cancer-driving kinases is an established strategy to improve the durability of clinical responses to targeted therapies. The difficulty of discovering kinase inhibitors with an appropriate multi-target profile has, however, necessitated the application of combination therapies, which can pose significant clinical development challenges. Epigenetic reader domains of the bromodomain family have recently emerged as novel targets for cancer therapy. Here we report that several clinical kinase inhibitors also inhibit bromodomains with therapeutically relevant potencies and are best classified as dual kinase/bromodomain inhibitors. Nanomolar activity on BRD4 by BI-2536 and TG-101348, clinical PLK1 and JAK2/FLT3 kinase inhibitors, respectively, is particularly noteworthy as these combinations of activities on independent oncogenic pathways exemplify a novel strategy for rational single agent polypharmacological targeting. Furthermore, structure-activity relationships and co-crystal structures identify design features that enable a general platform for the rational design of dual kinase/bromodomain inhibitors.


Journal of the American Chemical Society | 2014

Discovery and optimization of small-molecule ligands for the CBP/p300 bromodomains.

Duncan A. Hay; Oleg Fedorov; Sarah Martin; Dean C. Singleton; Cynthia Tallant; Christopher Wells; Sarah Picaud; Martin Philpott; Octovia P. Monteiro; Catherine Rogers; Stuart J. Conway; Timothy P. C. Rooney; Anthony Tumber; Clarence Yapp; Panagis Filippakopoulos; Mark Edward Bunnage; Susanne Müller; Stefan Knapp; Christopher J. Schofield; Paul E. Brennan

Small-molecule inhibitors that target bromodomains outside of the bromodomain and extra-terminal (BET) sub-family are lacking. Here, we describe highly potent and selective ligands for the bromodomain module of the human lysine acetyl transferase CBP/p300, developed from a series of 5-isoxazolyl-benzimidazoles. Our starting point was a fragment hit, which was optimized into a more potent and selective lead using parallel synthesis employing Suzuki couplings, benzimidazole-forming reactions, and reductive aminations. The selectivity of the lead compound against other bromodomain family members was investigated using a thermal stability assay, which revealed some inhibition of the structurally related BET family members. To address the BET selectivity issue, X-ray crystal structures of the lead compound bound to the CREB binding protein (CBP) and the first bromodomain of BRD4 (BRD4(1)) were used to guide the design of more selective compounds. The crystal structures obtained revealed two distinct binding modes. By varying the aryl substitution pattern and developing conformationally constrained analogues, selectivity for CBP over BRD4(1) was increased. The optimized compound is highly potent (Kd = 21 nM) and selective, displaying 40-fold selectivity over BRD4(1). Cellular activity was demonstrated using fluorescence recovery after photo-bleaching (FRAP) and a p53 reporter assay. The optimized compounds are cell-active and have nanomolar affinity for CBP/p300; therefore, they should be useful in studies investigating the biological roles of CBP and p300 and to validate the CBP and p300 bromodomains as therapeutic targets.


Journal of Medicinal Chemistry | 2013

Optimization of 3,5-dimethylisoxazole derivatives as potent bromodomain ligands.

David S. Hewings; Oleg Fedorov; Panagis Filippakopoulos; Sarah Martin; Sarah Picaud; Anthony Tumber; Christopher Wells; Monica M. Olcina; Katherine Freeman; Andrew Gill; Alison J. Ritchie; David W. Sheppard; Angela J. Russell; Ester M. Hammond; Stefan Knapp; Paul E. Brennan; Stuart J. Conway

The bromodomain protein module, which binds to acetylated lysine, is emerging as an important epigenetic therapeutic target. We report the structure-guided optimization of 3,5-dimethylisoxazole derivatives to develop potent inhibitors of the BET (bromodomain and extra terminal domain) bromodomain family with good ligand efficiency. X-ray crystal structures of the most potent compounds reveal key interactions required for high affinity at BRD4(1). Cellular studies demonstrate that the phenol and acetate derivatives of the lead compounds showed strong antiproliferative effects on MV4;11 acute myeloid leukemia cells, as shown for other BET bromodomain inhibitors and genetic BRD4 knockdown, whereas the reported compounds showed no general cytotoxicity in other cancer cell lines tested.


Cancer Research | 2015

Generation of a Selective Small Molecule Inhibitor of the CBP/p300 Bromodomain for Leukemia Therapy

Sarah Picaud; Oleg Fedorov; Angeliki Thanasopoulou; Katharina Leonards; Katherine Louise Jones; Julia Meier; Heidi Olzscha; Octovia P. Monteiro; Sarah Martin; Martin Philpott; Anthony Tumber; Panagis Filippakopoulos; Clarence Yapp; Christopher Wells; Ka Hing Che; Andrew J. Bannister; Samuel Robson; Umesh Kumar; Nigel James Parr; Kevin Lee; Dave Lugo; Philip Jeffrey; Simon Taylor; Matteo Vecellio; C. Bountra; Paul E. Brennan; Alison O'Mahony; Sharlene Velichko; Susanne Müller; Duncan Hay

The histone acetyltransferases CBP/p300 are involved in recurrent leukemia-associated chromosomal translocations and are key regulators of cell growth. Therefore, efforts to generate inhibitors of CBP/p300 are of clinical value. We developed a specific and potent acetyl-lysine competitive protein-protein interaction inhibitor, I-CBP112, that targets the CBP/p300 bromodomains. Exposure of human and mouse leukemic cell lines to I-CBP112 resulted in substantially impaired colony formation and induced cellular differentiation without significant cytotoxicity. I-CBP112 significantly reduced the leukemia-initiating potential of MLL-AF9(+) acute myeloid leukemia cells in a dose-dependent manner in vitro and in vivo. Interestingly, I-CBP112 increased the cytotoxic activity of BET bromodomain inhibitor JQ1 as well as doxorubicin. Collectively, we report the development and preclinical evaluation of a novel, potent inhibitor targeting CBP/p300 bromodomains that impairs aberrant self-renewal of leukemic cells. The synergistic effects of I-CBP112 and current standard therapy (doxorubicin) as well as emerging treatment strategies (BET inhibition) provide new opportunities for combinatorial treatment of leukemia and potentially other cancers.


Proceedings of the National Academy of Sciences of the United States of America | 2015

CBP30, a selective CBP/p300 bromodomain inhibitor, suppresses human Th17 responses

A Hammitzsch; Cynthia Tallant; Oleg Fedorov; Alison O’Mahony; Paul E. Brennan; Duncan A. Hay; Fernando O. Martinez; M. Hussein Al-Mossawi; Jelle de Wit; Matteo Vecellio; Christopher Wells; Paul Wordsworth; Susanne Müller; Stefan Knapp; Paul Bowness

Significance Epigenetic inhibitors have shown considerable promise for the treatment of malignant and inflammatory diseases. We present here the detailed characterization of a potent and highly selective inhibitor of the bromodomains of CBP (CREB binding protein)/p300. Functional preclinical data studying cells derived from patients with ankylosing spondylitis and psoriatic arthritis (two human Th17-driven diseases) show that selective inhibition of the CBP/p300 bromodomain with CBP30 strongly reduces secretion of IL-17A, without having the broader and potentially deleterious effects on cytokine production and gene transcription of the pan-BET (bromo and extraterminal domain protein family) inhibitor JQ1. CBP/p300 play a significant role in IL-17A production, and CBP/p300 inhibition is a promising therapeutic strategy in human type-17–mediated diseases such as ankylosing spondylitis and psoriatic arthritis. Th17 responses are critical to a variety of human autoimmune diseases, and therapeutic targeting with monoclonal antibodies against IL-17 and IL-23 has shown considerable promise. Here, we report data to support selective bromodomain blockade of the transcriptional coactivators CBP (CREB binding protein) and p300 as an alternative approach to inhibit human Th17 responses. We show that CBP30 has marked molecular specificity for the bromodomains of CBP and p300, compared with 43 other bromodomains. In unbiased cellular testing on a diverse panel of cultured primary human cells, CBP30 reduced immune cell production of IL-17A and other proinflammatory cytokines. CBP30 also inhibited IL-17A secretion by Th17 cells from healthy donors and patients with ankylosing spondylitis and psoriatic arthritis. Transcriptional profiling of human T cells after CBP30 treatment showed a much more restricted effect on gene expression than that observed with the pan-BET (bromo and extraterminal domain protein family) bromodomain inhibitor JQ1. This selective targeting of the CBP/p300 bromodomain by CBP30 will potentially lead to fewer side effects than with the broadly acting epigenetic inhibitors currently in clinical trials.


Science Advances | 2015

Selective targeting of the BRG/PB1 bromodomains impairs embryonic and trophoblast stem cell maintenance.

Oleg Fedorov; Josefina Castex; Cynthia Tallant; Dafydd R. Owen; Sarah Martin; Matteo Aldeghi; Octovia P. Monteiro; Panagis Filippakopoulos; Sarah Picaud; John David Trzupek; Brian S. Gerstenberger; C. Bountra; Dominica Willmann; Christopher Wells; Martin Philpott; Catherine Rogers; Philip C. Biggin; Paul E. Brennan; Mark Edward Bunnage; Roland Schüle; Thomas Günther; Stefan Knapp; Susanne Müller

PFI-3, a novel inhibitor targeting the bromodomains of essential components of the BAF/PBAF complex, affects the differentiation of ESC and TSC. Mammalian SWI/SNF [also called Brg/Brahma-associated factors (BAFs)] are evolutionarily conserved chromatin-remodeling complexes regulating gene transcription programs during development and stem cell differentiation. BAF complexes contain an ATP (adenosine 5′-triphosphate)–driven remodeling enzyme (either BRG1 or BRM) and multiple protein interaction domains including bromodomains, an evolutionary conserved acetyl lysine–dependent protein interaction motif that recruits transcriptional regulators to acetylated chromatin. We report a potent and cell active protein interaction inhibitor, PFI-3, that selectively binds to essential BAF bromodomains. The high specificity of PFI-3 was achieved on the basis of a novel binding mode of a salicylic acid head group that led to the replacement of water molecules typically maintained in other bromodomain inhibitor complexes. We show that exposure of embryonic stem cells to PFI-3 led to deprivation of stemness and deregulated lineage specification. Furthermore, differentiation of trophoblast stem cells in the presence of PFI-3 was markedly enhanced. The data present a key function of BAF bromodomains in stem cell maintenance and differentiation, introducing a novel versatile chemical probe for studies on acetylation-dependent cellular processes controlled by BAF remodeling complexes.


Science Advances | 2016

Promiscuous targeting of bromodomains by bromosporine identifies BET proteins as master regulators of primary transcription response in leukemia

Sarah Picaud; Katharina Leonards; Jean-Philippe Lambert; Oliver M. Dovey; Christopher Wells; Oleg Fedorov; Octovia P. Monteiro; Takao Fujisawa; Chen-Yi Wang; Hannah Lingard; Cynthia Tallant; Nikzad Nikbin; Lucie Guetzoyan; Richard J. Ingham; Steven V. Ley; Paul E. Brennan; Susanne Müller; Anastasia Samsonova; Anne-Claude Gingras; Juerg Schwaller; George S. Vassiliou; Stefan Knapp; Panagis Filippakopoulos

BET bromodomains are unique targeting modules that mediate primary transcription response. Bromodomains (BRDs) have emerged as compelling targets for cancer therapy. The development of selective and potent BET (bromo and extra-terminal) inhibitors and their significant activity in diverse tumor models have rapidly translated into clinical studies and have motivated drug development efforts targeting non-BET BRDs. However, the complex multidomain/subunit architecture of BRD protein complexes complicates predictions of the consequences of their pharmacological targeting. To address this issue, we developed a promiscuous BRD inhibitor [bromosporine (BSP)] that broadly targets BRDs (including BETs) with nanomolar affinity, creating a tool for the identification of cellular processes and diseases where BRDs have a regulatory function. As a proof of principle, we studied the effects of BSP on leukemic cell lines known to be sensitive to BET inhibition and found, as expected, strong antiproliferative activity. Comparison of the modulation of transcriptional profiles by BSP after a short exposure to the inhibitor resulted in a BET inhibitor signature but no significant additional changes in transcription that could account for inhibition of other BRDs. Thus, nonselective targeting of BRDs identified BETs, but not other BRDs, as master regulators of context-dependent primary transcription response.


Biology Open | 2013

Maintenance of muscle myosin levels in adult C. elegans requires both the double bromodomain protein BET-1 and sumoylation.

Kate Fisher; F Gee; S Wang; F Xue; Stefan Knapp; Martin Philpott; Christopher Wells; Miriam Rodriguez; L.B. Snoek; Jan E. Kammenga; Gino Poulin

Summary Attenuation of RAS-mediated signalling is a conserved process essential to control cell proliferation, differentiation, and apoptosis. Cooperative interactions between histone modifications such as acetylation, methylation and sumoylation are crucial for proper attenuation in C. elegans, implying that the proteins recognising these histone modifications could also play an important role in attenuation of RAS-mediated signalling. We sought to systematically identify these proteins and found BET-1. BET-1 is a conserved double bromodomain protein that recognises acetyl-lysines on histone tails and maintains the stable fate of various lineages. Unexpectedly, adults lacking both BET-1 and SUMO-1 are depleted of muscle myosin, an essential component of myofibrils. We also show that this muscle myosin depletion does not occur in all animals at a specific time, but rather that the penetrance of the phenotype increases with age. To gain mechanistic insights into this process, we sought to delay the occurrence of the muscle myosin depletion phenotype and found that it requires caspase activity and MEK-dependent signalling. We also performed transcription profiling on these mutants and found an up-regulation of the FGF receptor, egl-15, a tyrosine kinase receptor acting upstream of MEK. Consistent with a MEK requirement, we could delay the muscle phenotype by systemic or hypodermal knock down of egl-15. Thus, this work uncovered a caspase- and MEK-dependent mechanism that acts specifically on ageing adults to maintain the appropriate net level of muscle myosin.


Nature Chemical Biology | 2014

Corrigendum: Dual kinase-bromodomain inhibitors for rationally designed polypharmacology

Pietro Ciceri; Susanne Müller; Alison O'Mahony; Oleg Fedorov; Panagis Filippakopoulos; Jeremy P. Hunt; Elisabeth Lasater; Gabriel Pallares; Sarah Picaud; Christopher Wells; Sarah Martin; Lisa Wodicka; Neil P. Shah; Daniel Kelly Treiber; Stefan Knapp

Collaboration


Dive into the Christopher Wells's collaboration.

Top Co-Authors

Avatar

Stefan Knapp

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Neil P. Shah

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge