Nicholas A. Larsen
AstraZeneca
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Nicholas A. Larsen.
Structure | 2011
Andrew D. Ferguson; Nicholas A. Larsen; Tina Howard; Hannah Pollard; Isabelle Green; Christie Grande; Tony Cheung; Renee Garcia-Arenas; Scott Cowen; Jiaquan Wu; Robert Godin; Huawei Chen; Nicholas Keen
Protein lysine methyltransferases are important regulators of epigenetic signaling. These enzymes catalyze the transfer of donor methyl groups from S-adenosylmethionine to specific acceptor lysines on histones, leading to changes in chromatin structure and transcriptional regulation. These enzymes also methylate nonhistone protein substrates, revealing an additional mechanism to regulate cellular physiology. The oncogenic protein SMYD2 represses the functional activities of the tumor suppressor proteins p53 and Rb, making it an attractive drug target. Here we report the discovery of AZ505, a potent and selective inhibitor of SMYD2 that was identified from a high throughput chemical screen. We also present the crystal structures of SMYD2 with p53 substrate and product peptides, and notably, in complex with AZ505. This substrate competitive inhibitor is bound in the peptide binding groove of SMYD2. These results have implications for the development of SMYD2 inhibitors, and indicate the potential for developing novel therapies targeting this target class.
ACS Chemical Biology | 2013
Jiaquan Wu; Francoise Powell; Nicholas A. Larsen; Zhongwu Lai; Kate Byth; Jon Read; Rong-Fang Gu; Mark Roth; Dorin Toader; Jamal C. Saeh; Huawei Chen
Transforming growth factor-β activated kinase-1 (TAK1) is a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family that regulates several signaling pathways including NF-κB signal transduction and p38 activation. TAK1 deregulation has been implicated in human diseases including cancer and inflammation. Here, we show that, in addition to its kinase activity, TAK1 has intrinsic ATPase activity, that (5Z)-7-Oxozeaenol irreversibly inhibits TAK1, and that sensitivity to (5Z)-7-Oxozeaenol inhibition in hematological cancer cell lines is NRAS mutation status and TAK1 pathway dependent. X-ray crystallographic and mass spectrometric studies showed that (5Z)-7-Oxozeaenol forms a covalent complex with TAK1. Detailed biochemical characterization revealed that (5Z)-7-Oxozeaenol inhibited both the kinase and the ATPase activity of TAK1 following a bi-phase kinetics, consistent with the irreversible inhibition mechanism. In DoHH2 cells, (5Z)-7-Oxozeaenol potently inhibited the p38 phosphorylation driven by TAK1, and the inhibition lasted over 6 h after withdrawal of (5Z)-7-Oxozeaenol. Profiling (5Z)-7-Oxozeaenol in a panel of hematological cancer cells showed that sensitive cell lines tended to carry NRAS mutations and that genes in TAK1 regulated pathways were enriched in sensitive cell lines. Taken together, we have elucidated the molecular mechanism of a TAK1 irreversible inhibitor and laid the foundation for designing next generation TAK1 irreversible inhibitors. The NRAS-TAK1-Wnt signaling network discerned in our study may prove to be useful in patient selection for TAK1 targeted agents in hematological cancers.
Bioorganic & Medicinal Chemistry Letters | 2012
Tao Wang; Michael A. Block; Scott Cowen; Audrey Davies; Erik Devereaux; Lakshmaiah Gingipalli; Jeffrey W. Johannes; Nicholas A. Larsen; Qibin Su; Julie A. Tucker; David Whitston; Jiaquan Wu; Hai-Jun Zhang; Michael Zinda; Claudio Chuaqui
The design, synthesis and biological evaluation of a series of azabenzimidazole derivatives as TBK1/IKKε kinase inhibitors are described. Starting from a lead compound 1a, iterative design and SAR exploitation of the scaffold led to analogues with nM enzyme potencies against TBK1/IKKε. These compounds also exhibited excellent cellular activity against TBK1. Further structure-based design to improve selectivity over CDK2 and Aurora B resulted in compounds such as 5b-e. These probe compounds will facilitate study of the complex cancer biology of TBK1 and IKKε.
ACS Medicinal Chemistry Letters | 2013
James E. Dowling; Marat Alimzhanov; Larry Bao; Michael Howard Block; Claudio Chuaqui; Emma L. Cooke; Christopher R. Denz; Alex Hird; Shan Huang; Nicholas A. Larsen; Bo Peng; Timothy Pontz; Caroline Rivard-Costa; Jamal C. Saeh; Kumar Thakur; Qing Ye; Tao Zhang; Paul Lyne
In this letter, we describe the design, synthesis, and structure-activity relationship of 5-anilinopyrazolo[1,5-a]pyrimidine inhibitors of CK2 kinase. Property-based optimization of early leads using the 7-oxetan-3-yl amino group led to a series of matched molecular pairs with lower lipophilicity, decreased affinity for human plasma proteins, and reduced binding to the hERG ion channel. Agents in this study were shown to modulate pAKT(S129), a direct substrate of CK2, in vitro and in vivo, and exhibited tumor growth inhibition when administered orally in a murine DLD-1 xenograft.
Journal of Medicinal Chemistry | 2012
Gabriel Martinez-Botella; John N. Breen; James Duffy; Jacques Dumas; Bolin Geng; Ian K. Gowers; Oluyinka Green; Satenig Guler; Martin F. Hentemann; Felix A. Hernandez-Juan; Diane Joseph-McCarthy; Sameer Kawatkar; Nicholas A. Larsen; Ovadia Lazari; James T. Loch; Jacqueline Macritchie; Andrew R. McKenzie; Joseph V. Newman; Nelson B. Olivier; Linda G. Otterson; Andrew Pate Owens; Jon Read; David W. Sheppard; Thomas A. Keating
Thymidylate kinase (TMK) is an essential enzyme in bacterial DNA synthesis. The deoxythymidine monophosphate (dTMP) substrate binding pocket was targeted in a rational-design, structure-supported effort, yielding a unique series of antibacterial agents showing a novel, induced-fit binding mode. Lead optimization, aided by X-ray crystallography, led to picomolar inhibitors of both Streptococcus pneumoniae and Staphylococcus aureus TMK. MICs < 1 μg/mL were achieved against methicillin-resistant S. aureus (MRSA), S. pneumoniae, and vancomycin-resistant Enterococcus (VRE). Log D adjustments yielded single diastereomers 14 (TK-666) and 46, showing a broad antibacterial spectrum against Gram-positive bacteria and excellent selectivity against the human thymidylate kinase ortholog.
ACS Medicinal Chemistry Letters | 2012
James E. Dowling; Claudio Chuaqui; Timothy Pontz; Paul Lyne; Nicholas A. Larsen; Michael Howard Block; Huawei Chen; Nancy Su; Allan Wu; Daniel John Russell; Hannah Pollard; John W. Lee; Bo Peng; Kumar Thakur; Qing Ye; Tao Zhang; Patrick Brassil; Vicki Racicot; Larry Bao; Christopher R. Denz; Emma L. Cooke
In this paper we describe a series of 3-cyano-5-aryl-7-aminopyrazolo[1,5-a]pyrimidine hits identified by kinase-focused subset screening as starting points for the structure-based design of conformationally constrained 6-acetamido-indole inhibitors of CK2. The synthesis, SAR, and effects of this novel series on Akt signaling and cell proliferation in vitro are described.
Nature Communications | 2017
Teng Teng; Jennifer Tsai; Xiaoling Puyang; Michael Seiler; Shouyong Peng; Sudeep Prajapati; Daniel Aird; Silvia Buonamici; Benjamin Caleb; Betty Chan; Laura Corson; Jacob Feala; Peter Fekkes; Baudouin Gerard; Craig Karr; Manav Korpal; Xiang Liu; Jason T. Lowe; Yoshiharu Mizui; James Palacino; Eunice Park; P.G.R. Smith; V. Subramanian; Zhenhua Jeremy Wu; Jian Zou; Lihua Yu; Agustin Chicas; Markus Warmuth; Nicholas A. Larsen; Ping Zhu
Pladienolide, herboxidiene and spliceostatin have been identified as splicing modulators that target SF3B1 in the SF3b subcomplex. Here we report that PHF5A, another component of this subcomplex, is also targeted by these compounds. Mutations in PHF5A-Y36, SF3B1-K1071, SF3B1-R1074 and SF3B1-V1078 confer resistance to these modulators, suggesting a common interaction site. RNA-seq analysis reveals that PHF5A-Y36C has minimal effect on basal splicing but inhibits the global action of splicing modulators. Moreover, PHF5A-Y36C alters splicing modulator-induced intron-retention/exon-skipping profile, which correlates with the differential GC content between adjacent introns and exons. We determine the crystal structure of human PHF5A demonstrating that Y36 is located on a highly conserved surface. Analysis of the cryo-EM spliceosome Bact complex shows that the resistance mutations cluster in a pocket surrounding the branch point adenosine, suggesting a competitive mode of action. Collectively, we propose that PHF5A–SF3B1 forms a central node for binding to these splicing modulators.
Bioorganic & Medicinal Chemistry Letters | 2015
Jeffrey W. Johannes; Lynsie Almeida; Kevin Daly; Andrew D. Ferguson; Shaun Grosskurth; Huiping Guan; Tina Howard; Stephanos Ioannidis; Steven Kazmirski; Michelle Lamb; Nicholas A. Larsen; Paul Lyne; Keith Mikule; Claude Ogoe; Bo Peng; Philip Petteruti; Jon Read; Nancy Su; Mark Sylvester; Scott Throner; Wenxian Wang; Xin Wang; Jiaquan Wu; Qing Ye; Yan Yu; Xiaolan Zheng; David Scott
The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZenecas collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.
Bioorganic & Medicinal Chemistry Letters | 2012
Edward J. Hennessy; Jamal C. Saeh; Li Sha; Terry MacIntyre; Haiyun Wang; Nicholas A. Larsen; Brian Aquila; Andrew D. Ferguson; Naomi Laing; Charles A. Omer
A series of structurally unique Smac mimetics that act as antagonists of inhibitor of apoptosis proteins (IAPs) has been discovered. While most previously described Smac mimetics contain the proline ring (or a similar cyclic motif) found in Smac, a key feature of the compounds described herein is that this ring has been removed. Despite this, compounds in this series potently bind to cIAP1 and elicit the expected phenotype of cIAP1 inhibition in cancer cells. Marked selectivity for cIAP1 over XIAP is observed for these compounds, which is attributed to a slight difference in the binding groove between the two proteins and the resulting steric interactions with the inhibitors. XIAP binding can be improved by constraining the inhibitor so that these unfavorable steric interactions are minimized.
ACS Medicinal Chemistry Letters | 2016
James E. Dowling; Marat Alimzhanov; Larry Bao; Claudio Chuaqui; Christopher R. Denz; E Jenkins; Nicholas A. Larsen; Paul Lyne; Timothy Pontz; Qing Ye; G.A Holdgate; L Snow; N O'Connell; Andrew D. Ferguson
The Wnt pathway is an evolutionarily conserved and tightly regulated signaling network with important roles in embryonic development and adult tissue regeneration. Impaired Wnt pathway regulation, arising from mutations in Wnt signaling components, such as Axin, APC, and β-catenin, results in uncontrolled cell growth and triggers oncogenesis. To explore the reported link between CK2 kinase activity and Wnt pathway signaling, we sought to identify a potent, selective inhibitor of CK2 suitable for proof of concept studies in vivo. Starting from a pyrazolo[1,5-a]pyrimidine lead (2), we identified compound 7h, a potent CK2 inhibitor with picomolar affinity that is highly selectivity against other kinase family enzymes and inhibits Wnt pathway signaling (IC50 = 50 nM) in DLD-1 cells. In addition, compound 7h has physicochemical properties that are suitable for formulation as an intravenous solution, has demonstrated good pharmacokinetics in preclinical species, and exhibits a high level of activity as a monotherapy in HCT-116 and SW-620 xenografts.