41 results on '"Sharna J. Rich"'
Search Results
2. Supplementary Figure 3 from ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Gianni Chessari, Nicola G. Wallis, Nicola E. Wilsher, Pamela A. Williams, Neil T. Thompson, Tomoko Smyth, Sharna J. Rich, Joanne M. Munck, Vanessa Martins, John F. Lyons, Christopher N. Johnson, Jong Sook Ahn, Edward J. Lewis, and George A. Ward
- Abstract
XIAP antagonism measurements in HEK293-XIAP-Caspase-9 xenografts
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 2 from ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Gianni Chessari, Nicola G. Wallis, Nicola E. Wilsher, Pamela A. Williams, Neil T. Thompson, Tomoko Smyth, Sharna J. Rich, Joanne M. Munck, Vanessa Martins, John F. Lyons, Christopher N. Johnson, Jong Sook Ahn, Edward J. Lewis, and George A. Ward
- Abstract
XIAP antagonism in A375 cells in the presence or absence of TNF-α
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Materials and Methods from ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Gianni Chessari, Nicola G. Wallis, Nicola E. Wilsher, Pamela A. Williams, Neil T. Thompson, Tomoko Smyth, Sharna J. Rich, Joanne M. Munck, Vanessa Martins, John F. Lyons, Christopher N. Johnson, Jong Sook Ahn, Edward J. Lewis, and George A. Ward
- Abstract
Preparation of 2-chloro-1-{6-[(4-fluorophenyl)methyl]-5-(hydroxymethyl)-3,3-dimethyl-1H,2H,3H-pyrrolo[3,2-b]pyridin-1-yl}ethan-1-one
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Table 2 from ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Gianni Chessari, Nicola G. Wallis, Nicola E. Wilsher, Pamela A. Williams, Neil T. Thompson, Tomoko Smyth, Sharna J. Rich, Joanne M. Munck, Vanessa Martins, John F. Lyons, Christopher N. Johnson, Jong Sook Ahn, Edward J. Lewis, and George A. Ward
- Abstract
Fold induction in secreted cytokine concentrations in PBMC supernatants after IAP antagonist treatment
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figures 1-7 from ASTX029, a Novel Dual-mechanism ERK Inhibitor, Modulates Both the Phosphorylation and Catalytic Activity of ERK
- Author
-
Nicola G. Wallis, Alison J.-A. Woolford, Nicola E. Wilsher, Neil T. Thompson, Lukas Stanczuk, Alpesh D. Shah, Caroline J. Richardson, Sharna J. Rich, David C. Rees, Michael Reader, Marc O'Reilly, David Norton, Christopher W. Murray, Sandra Muench, Vanessa Martins, John F. Lyons, Justyna Kucia-Tran, Christopher J. Hindley, Tom D. Heightman, Roberta Ferraldeschi, Charlotte East, Aurélie Courtin, Juan Castro, Jessica L. Brothwood, Luke Bevan, Valerio Berdini, and Joanne M. Munck
- Abstract
Supplementary figures 1-7 show the effects of AST029 on ERK signalling in RAS-mutant cell lines (1) and tumour xenograft tissue (3), ASTX029 plasma PK linearity (2), ASTX029 activity in a cell line panel showing MAPK mutation status (4) the effects of ASTX029 on mouse body weight (5), PKPD effects of ASTX029 following b.i.d dosing to Colo205 tumour-bearing mice (6) and PD effects of ASTX029 following qd dosing to A375R tumour-bearing mice (7).
- Published
- 2023
- Full Text
- View/download PDF
7. Data from ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Gianni Chessari, Nicola G. Wallis, Nicola E. Wilsher, Pamela A. Williams, Neil T. Thompson, Tomoko Smyth, Sharna J. Rich, Joanne M. Munck, Vanessa Martins, John F. Lyons, Christopher N. Johnson, Jong Sook Ahn, Edward J. Lewis, and George A. Ward
- Abstract
Because of their roles in the evasion of apoptosis, inhibitor of apoptosis proteins (IAP) are considered attractive targets for anticancer therapy. Antagonists of these proteins have the potential to switch prosurvival signaling pathways in cancer cells toward cell death. Various SMAC-peptidomimetics with inherent cIAP selectivity have been tested clinically and demonstrated minimal single-agent efficacy. ASTX660 is a potent, non-peptidomimetic antagonist of cIAP1/2 and XIAP, discovered using fragment-based drug design. The antagonism of XIAP and cIAP1 by ASTX660 was demonstrated on purified proteins, cells, and in vivo in xenograft models. The compound binds to the isolated BIR3 domains of both XIAP and cIAP1 with nanomolar potencies. In cells and xenograft tissue, direct antagonism of XIAP was demonstrated by measuring its displacement from caspase-9 or SMAC. Compound-induced proteasomal degradation of cIAP1 and 2, resulting in downstream effects of NIK stabilization and activation of noncanonical NF-κB signaling, demonstrated cIAP1/2 antagonism. Treatment with ASTX660 led to TNFα-dependent induction of apoptosis in various cancer cell lines in vitro, whereas dosing in mice bearing breast and melanoma tumor xenografts inhibited tumor growth. ASTX660 is currently being tested in a phase I–II clinical trial (NCT02503423), and we propose that its antagonism of cIAP1/2 and XIAP may offer improved efficacy over first-generation antagonists that are more cIAP1/2 selective. Mol Cancer Ther; 17(7); 1381–91. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Table 1 from ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Gianni Chessari, Nicola G. Wallis, Nicola E. Wilsher, Pamela A. Williams, Neil T. Thompson, Tomoko Smyth, Sharna J. Rich, Joanne M. Munck, Vanessa Martins, John F. Lyons, Christopher N. Johnson, Jong Sook Ahn, Edward J. Lewis, and George A. Ward
- Abstract
Comparison of XIAP activity, cIAP1 activity and selectivity of clinical IAP antagonists
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 1 from ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Gianni Chessari, Nicola G. Wallis, Nicola E. Wilsher, Pamela A. Williams, Neil T. Thompson, Tomoko Smyth, Sharna J. Rich, Joanne M. Munck, Vanessa Martins, John F. Lyons, Christopher N. Johnson, Jong Sook Ahn, Edward J. Lewis, and George A. Ward
- Abstract
IncuCyte images from neutralization of TNF-α activity with anti-TNF-α experiment in MDA-MB-231 cells
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 4 from ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Gianni Chessari, Nicola G. Wallis, Nicola E. Wilsher, Pamela A. Williams, Neil T. Thompson, Tomoko Smyth, Sharna J. Rich, Joanne M. Munck, Vanessa Martins, John F. Lyons, Christopher N. Johnson, Jong Sook Ahn, Edward J. Lewis, and George A. Ward
- Abstract
Additional data from ASTX669 in vivo xenograft studies
- Published
- 2023
- Full Text
- View/download PDF
11. Fragment-Guided Discovery of Pyrazole Carboxylic Acid Inhibitors of the Kelch-like ECH-Associated Protein 1: Nuclear Factor Erythroid 2 Related Factor 2 (KEAP1:NRF2) Protein−Protein Interaction
- Author
-
Caroline Richardson, Zining Wu, Henriëtte M. G. Willems, William E. Wixted, Alison Jo-Anne Woolford, Charlotte Mary Griffiths-Jones, Yolanda Sanchez, David Norton, Jeffrey K. Kerns, T.G. Davies, William G. Bonnette, Sharna J. Rich, James F. Callahan, Hong Nie, Lawrence Wolfe, Maria Grazia Carr, Marcel L. Verdonk, William L. Rumsey, and Tom D. Heightman
- Subjects
chemistry.chemical_classification ,Kelch-Like ECH-Associated Protein 1 ,NF-E2-Related Factor 2 ,Drug discovery ,Carboxylic acid ,Carboxylic Acids ,Computational biology ,Pyrazole ,KEAP1 ,Keap1 nrf2 ,Cell Line ,Protein–protein interaction ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,chemistry ,Fragment (logic) ,Drug Discovery ,Animals ,Humans ,Pyrazoles ,Molecular Medicine ,Protein Binding - Abstract
The NRF2-mediated cytoprotective response is central to cellular homoeostasis, and there is increasing interest in developing small-molecule activators of this pathway as therapeutics for diseases involving chronic oxidative stress. The protein KEAP1, which regulates NRF2, is a key point for pharmacological intervention, and we recently described the use of fragment-based drug discovery to develop a tool compound that directly disrupts the protein-protein interaction between NRF2 and KEAP1. We now present the identification of a second, chemically distinct series of KEAP1 inhibitors, which provided an alternative chemotype for lead optimization. Pharmacophoric information from our original fragment screen was used to identify new hit matter through database searching and to evolve this into a new lead with high target affinity and cell-based activity. We highlight how knowledge obtained from fragment-based approaches can be used to focus additional screening campaigns in order to de-risk projects through the rapid identification of novel chemical series.
- Published
- 2021
- Full Text
- View/download PDF
12. ASTX029, a Novel Dual-mechanism ERK Inhibitor, Modulates Both the Phosphorylation and Catalytic Activity of ERK
- Author
-
Neil T. Thompson, Nicola G. Wallis, Michael Reader, Christopher J. Hindley, Juan Castro, John Lyons, David C. Rees, Nicola E. Wilsher, Caroline Richardson, Tom D. Heightman, Lukas Stanczuk, Alpesh Shah, Sharna J. Rich, Charlotte East, Valerio Berdini, Marc O'Reilly, Justyna Kucia-Tran, Jessica L. Brothwood, Joanne M. Munck, Alison Jo-Anne Woolford, Sandra Muench, Christopher William Murray, David Norton, Luke Bevan, Aurélie Courtin, Roberta Ferraldeschi, and Vanessa Martins
- Subjects
Male ,MAPK/ERK pathway ,Cancer Research ,Indoles ,Mice, Nude ,Apoptosis ,Ribosomal s6 kinase ,Mice ,Downregulation and upregulation ,Cell Movement ,In vivo ,Tumor Cells, Cultured ,Animals ,Humans ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Protein Kinase Inhibitors ,Cell Proliferation ,Mice, Inbred BALB C ,biology ,Chemistry ,Effector ,Cell Cycle ,Xenograft Model Antitumor Assays ,In vitro ,Gene Expression Regulation, Neoplastic ,Pyrimidines ,Oncology ,Drug Resistance, Neoplasm ,Cell culture ,Colonic Neoplasms ,Cancer research ,biology.protein - Abstract
The MAPK signaling pathway is commonly upregulated in human cancers. As the primary downstream effector of the MAPK pathway, ERK is an attractive therapeutic target for the treatment of MAPK-activated cancers and for overcoming resistance to upstream inhibition. ASTX029 is a highly potent and selective dual-mechanism ERK inhibitor, discovered using fragment-based drug design. Because of its distinctive ERK-binding mode, ASTX029 inhibits both ERK catalytic activity and the phosphorylation of ERK itself by MEK, despite not directly inhibiting MEK activity. This dual mechanism was demonstrated in cell-free systems, as well as cell lines and xenograft tumor tissue, where the phosphorylation of both ERK and its substrate, ribosomal S6 kinase (RSK), were modulated on treatment with ASTX029. Markers of sensitivity were highlighted in a large cell panel, where ASTX029 preferentially inhibited the proliferation of MAPK-activated cell lines, including those with BRAF or RAS mutations. In vivo, significant antitumor activity was observed in MAPK-activated tumor xenograft models following oral treatment. ASTX029 also demonstrated activity in both in vitro and in vivo models of acquired resistance to MAPK pathway inhibitors. Overall, these findings highlight the therapeutic potential of a dual-mechanism ERK inhibitor such as ASTX029 for the treatment of MAPK-activated cancers, including those which have acquired resistance to inhibitors of upstream components of the MAPK pathway. ASTX029 is currently being evaluated in a first in human phase I–II clinical trial in patients with advanced solid tumors (NCT03520075).
- Published
- 2021
- Full Text
- View/download PDF
13. Discovery of ASTX029, A Clinical Candidate Which Modulates the Phosphorylation and Catalytic Activity of ERK1/2
- Author
-
Michael Reader, Aurélie Courtin, Steven Howard, Nicola G. Wallis, Jonathan Shannon, Hugh Walton, Christopher William Murray, Joseph E. Coyle, Joanne M. Munck, Alison Jo-Anne Woolford, Torren M. Peakman, David C. Rees, James Edward Harvey Day, Charlotte Mary Griffiths-Jones, Marc O'Reilly, Alpesh Shah, David Norton, Valerio Berdini, Lynsey Fazal, Maria Grazia Carr, Charlotte East, Tom D. Heightman, Nicola E. Wilsher, Ildiko Maria Buck, Luke Bevan, Stuart Thomas Onions, Michael Cooke, Justyna Kucia-Tran, Sandra Muench, Nick Palmer, Sharna J. Rich, Vanessa Martins, David Cousin, Puja Pathuri, and John P. Watts
- Subjects
Male ,Indoles ,Antineoplastic Agents ,Crystallography, X-Ray ,Proto-Oncogene Mas ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,Dogs ,Pharmacokinetics ,Neoplasms ,Drug Discovery ,Animals ,Humans ,In patient ,Tumor growth ,Phosphorylation ,Rats, Wistar ,Protein kinase A ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase 1 ,Mice, Inbred BALB C ,Molecular Structure ,Chemistry ,Kinase ,Xenograft Model Antitumor Assays ,Clinical trial ,Pyrimidines ,Cancer research ,Molecular Medicine ,Once daily dosing ,Protein Binding - Abstract
Aberrant activation of the mitogen-activated protein kinase pathway frequently drives tumor growth, and the ERK1/2 kinases are positioned at a key node in this pathway, making them important targets for therapeutic intervention. Recently, a number of ERK1/2 inhibitors have been advanced to investigational clinical trials in patients with activating mutations in B-Raf proto-oncogene or Ras. Here, we describe the discovery of the clinical candidate ASTX029 (15) through structure-guided optimization of our previously published isoindolinone lead (7). The medicinal chemistry campaign focused on addressing CYP3A4-mediated metabolism and maintaining favorable physicochemical properties. These efforts led to the identification of ASTX029, which showed the desired pharmacological profile combining ERK1/2 inhibition with suppression of phospho-ERK1/2 (pERK) levels, and in addition, it possesses suitable preclinical pharmacokinetic properties predictive of once daily dosing in humans. ASTX029 is currently in a phase I-II clinical trial in patients with advanced solid tumors.
- Published
- 2021
14. A Fragment-Derived Clinical Candidate for Antagonism of X-Linked and Cellular Inhibitor of Apoptosis Proteins: 1-(6-[(4-Fluorophenyl)methyl]-5-(hydroxymethyl)-3,3-dimethyl-1H,2H,3H-pyrrolo[3,2-b]pyridin-1-yl)-2-[(2R,5R)-5-methyl-2-([(3R)-3-methylmorpholin-4-yl]methyl)piperazin-1-yl]ethan-1-one (ASTX660)
- Author
-
Gianni Chessari, Ildiko Maria Buck, Edward J. Lewis, Elisabetta Chiarparin, Jong Sook Ahn, Gordon Saxty, Anna Hopkins, Nicola E. Wilsher, Michael Reader, George Ward, Torren M. Peakman, Steven Howard, Pamela A. Williams, Tomoko Smyth, Christopher N. Johnson, Neil T. Thompson, Vanessa Martins, Joanne M. Munck, Alessia Millemaggi, James Edward Harvey Day, Lee William Page, and Sharna J. Rich
- Subjects
010405 organic chemistry ,Peptidomimetic ,Stereochemistry ,Antagonist ,Inhibitor of apoptosis ,01 natural sciences ,0104 chemical sciences ,XIAP ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,Apoptosis ,030220 oncology & carcinogenesis ,Drug Discovery ,Molecular Medicine ,Hydroxymethyl ,Selectivity ,Antagonism - Abstract
Inhibitor of apoptosis proteins (IAPs) are promising anticancer targets, given their roles in the evasion of apoptosis. Several peptidomimetic IAP antagonists, with inherent selectivity for cellular IAP (cIAP) over X-linked IAP (XIAP), have been tested in the clinic. A fragment screening approach followed by structure-based optimization has previously been reported that resulted in a low-nanomolar cIAP1 and XIAP antagonist lead molecule with a more balanced cIAP–XIAP profile. We now report the further structure-guided optimization of the lead, with a view to improving the metabolic stability and cardiac safety profile, to give the nonpeptidomimetic antagonist clinical candidate 27 (ASTX660), currently being tested in a phase 1/2 clinical trial (NCT02503423).
- Published
- 2018
- Full Text
- View/download PDF
15. ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFα-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth
- Author
-
Pamela A. Williams, Tomoko Smyth, Christopher N. Johnson, Edward J. Lewis, Vanessa Martins, Nicola G. Wallis, John Lyons, Jong Sook Ahn, Nicola E. Wilsher, Joanne M. Munck, Neil T. Thompson, Gianni Chessari, Sharna J. Rich, and George Ward
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Antineoplastic Agents ,Apoptosis ,X-Linked Inhibitor of Apoptosis Protein ,Inhibitor of apoptosis ,Inhibitor of Apoptosis Proteins ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Cell Proliferation ,Dose-Response Relationship, Drug ,Tumor Necrosis Factor-alpha ,Chemistry ,Molecular Mimicry ,Xenograft Model Antitumor Assays ,XIAP ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Signal transduction ,Antagonism - Abstract
Because of their roles in the evasion of apoptosis, inhibitor of apoptosis proteins (IAP) are considered attractive targets for anticancer therapy. Antagonists of these proteins have the potential to switch prosurvival signaling pathways in cancer cells toward cell death. Various SMAC-peptidomimetics with inherent cIAP selectivity have been tested clinically and demonstrated minimal single-agent efficacy. ASTX660 is a potent, non-peptidomimetic antagonist of cIAP1/2 and XIAP, discovered using fragment-based drug design. The antagonism of XIAP and cIAP1 by ASTX660 was demonstrated on purified proteins, cells, and in vivo in xenograft models. The compound binds to the isolated BIR3 domains of both XIAP and cIAP1 with nanomolar potencies. In cells and xenograft tissue, direct antagonism of XIAP was demonstrated by measuring its displacement from caspase-9 or SMAC. Compound-induced proteasomal degradation of cIAP1 and 2, resulting in downstream effects of NIK stabilization and activation of noncanonical NF-κB signaling, demonstrated cIAP1/2 antagonism. Treatment with ASTX660 led to TNFα-dependent induction of apoptosis in various cancer cell lines in vitro, whereas dosing in mice bearing breast and melanoma tumor xenografts inhibited tumor growth. ASTX660 is currently being tested in a phase I–II clinical trial (NCT02503423), and we propose that its antagonism of cIAP1/2 and XIAP may offer improved efficacy over first-generation antagonists that are more cIAP1/2 selective. Mol Cancer Ther; 17(7); 1381–91. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
16. Fragment-Based Discovery of a Potent, Orally Bioavailable Inhibitor That Modulates the Phosphorylation and Catalytic Activity of ERK1/2
- Author
-
Neil T. Thompson, Charlotte Mary Griffiths-Jones, John Lyons, Vanessa Martins, Nicola E. Wilsher, Tom D. Heightman, Harpreet K Saini, Juan Castro, Stuart Thomas Onions, David Cousin, Nicola G. Wallis, Valerio Berdini, Nick Palmer, Puja Pathuri, Caroline Richardson, Christopher William Murray, Charlotte East, John P. Watts, Marc O'Reilly, Sharna J. Rich, Hugh Walton, James Edward Harvey Day, Aurélie Courtin, Brent Graham, David C. Rees, Hannah Braithwaite, Michael Cooke, Ildiko Maria Buck, Michael Reader, Sandra Muench, Megan Cassidy, Jonathan Shannon, Joanne M. Munck, Alison Jo-Anne Woolford, David Norton, and Lynsey Fazal
- Subjects
Models, Molecular ,0301 basic medicine ,MAPK/ERK pathway ,Protein Conformation ,Mutant ,Administration, Oral ,Biological Availability ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Drug Discovery ,medicine ,Animals ,Humans ,Kinome ,Phosphorylation ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Drug discovery ,Chemistry ,Cell growth ,Melanoma ,medicine.disease ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,Biocatalysis ,Cancer research ,Molecular Medicine - Abstract
Aberrant activation of the MAPK pathway drives cell proliferation in multiple cancers. Inhibitors of BRAF and MEK kinases are approved for the treatment of BRAF mutant melanoma, but resistance frequently emerges, often mediated by increased signaling through ERK1/2. Here, we describe the fragment-based generation of ERK1/2 inhibitors that block catalytic phosphorylation of downstream substrates such as RSK but also modulate phosphorylation of ERK1/2 by MEK without directly inhibiting MEK. X-ray crystallographic and biophysical fragment screening followed by structure-guided optimization and growth from the hinge into a pocket proximal to the C-α helix afforded highly potent ERK1/2 inhibitors with excellent kinome selectivity. In BRAF mutant cells, the lead compound suppresses pRSK and pERK levels and inhibits proliferation at low nanomolar concentrations. The lead exhibits tumor regression upon oral dosing in BRAF mutant xenograft models, providing a promising basis for further optimization toward clinical pERK1/2 modulating ERK1/2 inhibitors.
- Published
- 2018
- Full Text
- View/download PDF
17. Discovery of a Potent Nonpeptidomimetic, Small-Molecule Antagonist of Cellular Inhibitor of Apoptosis Protein 1 (cIAP1) and X-Linked Inhibitor of Apoptosis Protein (XIAP)
- Author
-
Christopher N. Johnson, Nicola E. Wilsher, Vanessa Martins, Gianni Chessari, Aman Iqbal, Torren M. Peakman, Pamela A. Williams, Keisha Hearn, George Ward, Edward J. Lewis, James Edward Harvey Day, Ildiko Maria Buck, Charlotte Mary Griffiths-Jones, Tom D. Heightman, Michael Reader, Sharna J. Rich, Martyn Frederickson, Elisabetta Chiarparin, and Emiliano Tamanini
- Subjects
0301 basic medicine ,X-Linked Inhibitor of Apoptosis Protein ,Mice, SCID ,Crystallography, X-Ray ,Inhibitor of apoptosis ,Heterocyclic Compounds, 2-Ring ,Piperazines ,Inhibitor of Apoptosis Proteins ,Small Molecule Libraries ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Inhibitor of apoptosis domain ,Mice, Inbred BALB C ,Drug discovery ,Chemistry ,Small molecule ,XIAP ,Cell biology ,body regions ,HEK293 Cells ,030104 developmental biology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Peptidomimetics ,Baculoviral IAP repeat-containing protein 3 ,Signal transduction - Abstract
XIAP and cIAP1 are members of the inhibitor of apoptosis protein (IAP) family and are key regulators of anti-apoptotic and pro-survival signaling pathways. Overexpression of IAPs occurs in various cancers and has been associated with tumor progression and resistance to treatment. Structure-based drug design (SBDD) guided by structural information from X-ray crystallography, computational studies, and NMR solution conformational analysis was successfully applied to a fragment-derived lead resulting in AT-IAP, a potent, orally bioavailable, dual antagonist of XIAP and cIAP1 and a structurally novel chemical probe for IAP biology.
- Published
- 2017
- Full Text
- View/download PDF
18. Fragment-Based Approach to the Development of an Orally Bioavailable Lactam Inhibitor of Lipoprotein-Associated Phospholipase A2 (Lp-PLA2)
- Author
-
Yann Dudit, Zehong Wan, Don O. Somers, Yingxia Sang, Vipulkumar Kantibhai Patel, Pascal Grondin, Joseph E. Coyle, Veronique Beneton, Rachel McMenamin, Lee William Page, Philip J. Day, Alison Jo-Anne Woolford, Lionel Trottet, Valerio Berdini, Florent Potvain, Sharna J. Rich, Christopher William Murray, Pascal Huet, Lydia Y. W. Lee, Xiaomin Zhang, and Eric S. Manas
- Subjects
0301 basic medicine ,Virtual screening ,biology ,Lipoprotein-associated phospholipase A2 ,Active site ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Fragment (logic) ,030220 oncology & carcinogenesis ,Drug Discovery ,Lipophilicity ,Lactam ,biology.protein ,Molecular Medicine ,Bioassay ,lipids (amino acids, peptides, and proteins) ,IC50 - Abstract
Lp-PLA2 has been explored as a target for a number of inflammation associated diseases, including cardiovascular disease and dementia. This article describes the discovery of a new fragment derived chemotype that interacts with the active site of Lp-PLA2. The starting fragment hit was discovered through an X-ray fragment screen and showed no activity in the bioassay (IC50 > 1 mM). The fragment hit was optimized using a variety of structure-based drug design techniques, including virtual screening, fragment merging, and improvement of shape complementarity. A novel series of Lp-PLA2 inhibitors was generated with low lipophilicity and a promising pharmacokinetic profile.
- Published
- 2016
- Full Text
- View/download PDF
19. Structure of the Epigenetic Oncogene MMSET and Inhibition by N-Alkyl Sinefungin Derivatives
- Author
-
Emiliano Tamanini, Joseph E. Coyle, Agnes C. L. Martin, Adam Hold, Elisabetta Chiarparin, Sharna J. Rich, Ludovic Drouin, Puja Pathuri, Rob L. M. van Montfort, Tom D. Heightman, Marcello Tortorici, Finn P. Holding, Jeff Yon, Amy Wood, Rosemary Burke, Fiona Jeganathan, Yvette Newbatt, Nader Amin, Jenny E. Harmer, Maura Westlake, Paul Acklam, Swen Hoelder, Valerio Berdini, and Dominic Tisi
- Subjects
0301 basic medicine ,Adenosine ,Magnetic Resonance Spectroscopy ,Protein Conformation ,Stereochemistry ,Mutant ,Calorimetry ,Biology ,Crystallography, X-Ray ,Biochemistry ,Mass Spectrometry ,Epigenesis, Genetic ,03 medical and health sciences ,Sinefungin ,Protein structure ,SETD2 ,Transferase ,Binding site ,Binding Sites ,Point mutation ,Histone-Lysine N-Methyltransferase ,Oncogenes ,General Medicine ,Small molecule ,Repressor Proteins ,030104 developmental biology ,Drug Design ,Molecular Medicine ,Chromatography, Liquid - Abstract
The members of the NSD subfamily of lysine methyl transferases are compelling oncology targets due to the recent characterization of gain-of-function mutations and translocations in several hematological cancers. To date, these proteins have proven intractable to small molecule inhibition. Here, we present initial efforts to identify inhibitors of MMSET (aka NSD2 or WHSC1) using solution phase and crystal structural methods. On the basis of 2D NMR experiments comparing NSD1 and MMSET structural mobility, we designed an MMSET construct with five point mutations in the N-terminal helix of its SET domain for crystallization experiments and elucidated the structure of the mutant MMSET SET domain at 2.1 Å resolution. Both NSD1 and MMSET crystal systems proved resistant to soaking or cocrystallography with inhibitors. However, use of the close homologue SETD2 as a structural surrogate supported the design and characterization of N-alkyl sinefungin derivatives, which showed low micromolar inhibition against both SETD2 and MMSET.
- Published
- 2016
- Full Text
- View/download PDF
20. Exploitation of a Novel Binding Pocket in Human Lipoprotein-Associated Phospholipase A2 (Lp-PLA2) Discovered through X-ray Fragment Screening
- Author
-
Kirsten V. Smith, Joseph E. Pero, Ralph A. Rivero, Donald O. Somers, Vipulkumar Kantibhai Patel, Agnes C. L. Martin, Rachel McMenamin, Sridhar Aravapalli, Finn P. Holding, Eric S. Manas, Lionel Trottet, Alison Jo-Anne Woolford, Neipp Christopher E, Lydia Y. W. Lee, Ranganadh Velagaleti, Sharna J. Rich, Ren Xie, Peng Li, Glyn Williams, Andrew M. Dodson, Brent W. Mccleland, Lee William Page, Joseph P. Marino, Pascal Grondin, Florent Potvain, Joseph E. Coyle, Christopher William Murray, Philip J. Day, and Valerio Berdini
- Subjects
Models, Molecular ,0301 basic medicine ,Stereochemistry ,Drug Evaluation, Preclinical ,Crystallography, X-Ray ,Structure-Activity Relationship ,03 medical and health sciences ,Phospholipase A2 ,Drug Discovery ,Hydrolase ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,Binding site ,Binding Sites ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Chemistry ,Drug discovery ,Lipoprotein-associated phospholipase A2 ,Active site ,1-Alkyl-2-acetylglycerophosphocholine Esterase ,Thiazoles ,030104 developmental biology ,Biochemistry ,biology.protein ,Pyrazoles ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) - Abstract
Elevated levels of human lipoprotein-associated phospholipase A2 (Lp-PLA2) are associated with cardiovascular disease and dementia. A fragment screen was conducted against Lp-PLA2 in order to identify novel inhibitors. Multiple fragment hits were observed in different regions of the active site, including some hits that bound in a pocket created by movement of a protein side chain (approximately 13 Å from the catalytic residue Ser273). Using structure guided design, we optimized a fragment that bound in this pocket to generate a novel low nanomolar chemotype, which did not interact with the catalytic residues.
- Published
- 2016
- Full Text
- View/download PDF
21. Identification of potent small molecule allosteric inhibitors of SHP2
- Author
-
Nicola G. Wallis, Valerio Berdini, S. Hiscock, T.G. Davies, H. Ochiiwa, Keisha Hearn, Y. Nakatsuru, Tomoko Smyth, Sharna J. Rich, Sandra Muench, Alpesh Shah, A. Price, Y. Shibata, T. Shimamura, Nicola E. Wilsher, C.N. Johnson, G. Chessari, Vanessa Martins, J.E.H. Day, and C. Hamlett
- Subjects
Cancer Research ,Oncology ,Biochemistry ,Chemistry ,Allosteric regulation ,Identification (biology) ,Small molecule - Published
- 2020
- Full Text
- View/download PDF
22. The clinical candidate, ASTX029, is a novel, dual mechanism ERK1/2 inhibitor and has potent activity in MAPK-activated cancer cell lines and in vivo tumor models
- Author
-
Nicola G. Wallis, Christopher J. Hindley, D. Norton, C. East, Vanessa Martins, John C. Lyons, Nicola E. Wilsher, Joanne M. Munck, Valerio Berdini, Aurélie Courtin, Sharna J. Rich, David C. Rees, J. Kucia-Tran, Neil T. Thompson, T. Heightman, A.J.-A. Woolford, Christopher W. Murray, Sandra Muench, M. Reader, and M. O’Reilly
- Subjects
MAPK/ERK pathway ,Cancer Research ,Oncology ,In vivo ,Chemistry ,Cancer research ,Cancer cell lines ,Dual mechanism - Published
- 2020
- Full Text
- View/download PDF
23. A Fragment-Derived Clinical Candidate for Antagonism of X-Linked and Cellular Inhibitor of Apoptosis Proteins: 1-(6-[(4-Fluorophenyl)methyl]-5-(hydroxymethyl)-3,3-dimethyl-1 H,2 H,3 H-pyrrolo[3,2- b]pyridin-1-yl)-2-[(2 R,5 R)-5-methyl-2-([(3R)-3-methylmorpholin-4-yl]methyl)piperazin-1-yl]ethan-1-one (ASTX660)
- Author
-
Christopher N, Johnson, Jong Sook, Ahn, Ildiko M, Buck, Elisabetta, Chiarparin, James E H, Day, Anna, Hopkins, Steven, Howard, Edward J, Lewis, Vanessa, Martins, Alessia, Millemaggi, Joanne M, Munck, Lee W, Page, Torren, Peakman, Michael, Reader, Sharna J, Rich, Gordon, Saxty, Tomoko, Smyth, Neil T, Thompson, George A, Ward, Pamela A, Williams, Nicola E, Wilsher, and Gianni, Chessari
- Subjects
Male ,ERG1 Potassium Channel ,Mice, Inbred BALB C ,Administration, Oral ,Antineoplastic Agents ,X-Linked Inhibitor of Apoptosis Protein ,Crystallography, X-Ray ,Heterocyclic Compounds, 2-Ring ,Xenograft Model Antitumor Assays ,Piperazines ,Inhibitor of Apoptosis Proteins ,Rats, Sprague-Dawley ,Macaca fascicularis ,Structure-Activity Relationship ,Cell Line, Tumor ,Animals ,Humans - Abstract
Inhibitor of apoptosis proteins (IAPs) are promising anticancer targets, given their roles in the evasion of apoptosis. Several peptidomimetic IAP antagonists, with inherent selectivity for cellular IAP (cIAP) over X-linked IAP (XIAP), have been tested in the clinic. A fragment screening approach followed by structure-based optimization has previously been reported that resulted in a low-nanomolar cIAP1 and XIAP antagonist lead molecule with a more balanced cIAP-XIAP profile. We now report the further structure-guided optimization of the lead, with a view to improving the metabolic stability and cardiac safety profile, to give the nonpeptidomimetic antagonist clinical candidate 27 (ASTX660), currently being tested in a phase 1/2 clinical trial (NCT02503423).
- Published
- 2018
24. Crystal structure of human soluble adenylate cyclase reveals a distinct, highly flexible allosteric bicarbonate binding pocket
- Author
-
Christopher William Murray, Joseph E. Coyle, Sharna J. Rich, Anne Cleasby, Tracey Ann Sambrook, S. Saalau-Bethell, Jeff Yon, Valerio Berdini, Mladen Vinkovic, Victoria Lock, M. Alistair O’Brien, Miles Congreve, and Harren Jhoti
- Subjects
Models, Molecular ,Stereochemistry ,Allosteric regulation ,Adenylate kinase ,Crystallography, X-Ray ,Biochemistry ,Cyclase ,drug discovery ,Enzyme activator ,Structure-Activity Relationship ,Catalytic Domain ,Humans ,structural biology ,General Pharmacology, Toxicology and Pharmaceutics ,Enzyme Inhibitors ,Bicarbonate binding ,Pharmacology ,chemistry.chemical_classification ,allosterism ,biology ,Dose-Response Relationship, Drug ,Organic Chemistry ,Active site ,fragment screening ,Full Papers ,Enzyme Activation ,Bicarbonates ,Enzyme ,chemistry ,Allosteric enzyme ,Adenylyl Cyclase Inhibitors ,biology.protein ,Molecular Medicine ,enzyme regulation ,Adenylyl Cyclases - Abstract
Soluble adenylate cyclases catalyse the synthesis of the second messenger cAMP through the cyclisation of ATP and are the only known enzymes to be directly activated by bicarbonate. Here, we report the first crystal structure of the human enzyme that reveals a pseudosymmetrical arrangement of two catalytic domains to produce a single competent active site and a novel discrete bicarbonate binding pocket. Crystal structures of the apo protein, the protein in complex with α,β-methylene adenosine 5′-triphosphate (AMPCPP) and calcium, with the allosteric activator bicarbonate, and also with a number of inhibitors identified using fragment screening, all show a flexible active site that undergoes significant conformational changes on binding of ligands. The resulting nanomolar-potent inhibitors that were developed bind at both the substrate binding pocket and the allosteric site, and can be used as chemical probes to further elucidate the function of this protein.
- Published
- 2014
25. Fragment-Based Approach to the Development of an Orally Bioavailable Lactam Inhibitor of Lipoprotein-Associated Phospholipase A2 (Lp-PLA
- Author
-
Alison J-A, Woolford, Philip J, Day, Véronique, Bénéton, Valerio, Berdini, Joseph E, Coyle, Yann, Dudit, Pascal, Grondin, Pascal, Huet, Lydia Y W, Lee, Eric S, Manas, Rachel L, McMenamin, Christopher W, Murray, Lee W, Page, Vipulkumar K, Patel, Florent, Potvain, Sharna J, Rich, Yingxia, Sang, Don O, Somers, Lionel, Trottet, Zehong, Wan, and Xiaomin, Zhang
- Subjects
Models, Molecular ,Dose-Response Relationship, Drug ,Lactams ,Molecular Structure ,Administration, Oral ,Biological Availability ,Crystallography, X-Ray ,Rats ,Structure-Activity Relationship ,Dogs ,1-Alkyl-2-acetylglycerophosphocholine Esterase ,Animals ,Humans ,Tissue Distribution ,Enzyme Inhibitors - Abstract
Lp-PLA
- Published
- 2016
26. Discovery of an allosteric mechanism for the regulation of HCV NS3 protein function
- Author
-
Pamela A. Williams, Harren Jhoti, Andrew James Woodhead, Maria Grazia Carr, Puja Pathuri, Christopher William Murray, Caroline Richardson, Brent Graham, S. Saalau-Bethell, Gianni Chessari, Steven Douglas Hiscock, Joseph T. Coyle, and Sharna J. Rich
- Subjects
Models, Molecular ,NS3 ,Protease ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,medicine.medical_treatment ,Protein domain ,Allosteric regulation ,Helicase ,Cell Biology ,Viral Nonstructural Proteins ,Ligands ,Antiviral Agents ,Article ,NS2-3 protease ,Structure-Activity Relationship ,Allosteric Regulation ,Biochemistry ,medicine ,biology.protein ,Structure–activity relationship ,Binding site ,Molecular Biology ,Allosteric Site - Abstract
Here we report the discovery of a highly conserved novel binding site located at the interface between the protease and helicase domains of the Hepatitis C Virus (HCV) NS3 protein. Using a chemical lead, identified by fragment screening and structure-guided design, we demonstrate that this site has a regulatory function on the protease activity via an allosteric mechanism. We propose that compounds binding at this allosteric site inhibit the function of the NS3 protein by stabilising an inactive conformation and thus represent a new class of direct acting antiviral agents.
- Published
- 2012
- Full Text
- View/download PDF
27. Potent, Selective Inhibitors of Fibroblast Growth Factor Receptor Define Fibroblast Growth Factor Dependence in Preclinical Cancer Models
- Author
-
Martyn Frederickson, Christopher W. Murray, Neil T. Thompson, Tim Perera, Anne Cleasby, Eddy Jean Edgard Freyne, Ruth Feltell, Jose Cosme, Lynsey Fazal, Christopher Charles Frederick Hamlett, Michael Reader, Rajdeep Benning, Alistair O'Brien, Peter King, Darcey Miller, Matthew Squires, Patrick Angibaud, Andrew Madin, Valerio Berdini, Emma Vickerstaffe, Gordon Saxty, Brent Graham, Charlotte Griffiths Jones, Andrew Pike, Edward J. Lewis, Michael A. Batey, Douglas D. Ross, Susanne S. Bethell, Sharna J. Rich, Lindsay A. Devine, Maria Grazia Carr, Abarna Thiru, Julie Irving, George Ward, John Lyons, and David R. Newell
- Subjects
Models, Molecular ,musculoskeletal diseases ,Gene isoform ,Cancer Research ,animal structures ,Cell Survival ,Drug Evaluation, Preclinical ,Mice, Nude ,Antineoplastic Agents ,Fibroblast growth factor ,Receptor tyrosine kinase ,Mice ,In vivo ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Receptor ,Protein Kinase Inhibitors ,Cell Proliferation ,Mice, Inbred BALB C ,biology ,Chemistry ,Cancer ,medicine.disease ,Receptors, Fibroblast Growth Factor ,Xenograft Model Antitumor Assays ,Fibroblast Growth Factors ,Treatment Outcome ,Oncology ,Cell culture ,Fibroblast growth factor receptor ,Drug Design ,embryonic structures ,Immunology ,biology.protein ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Signal Transduction - Abstract
We describe here the identification and characterization of 2 novel inhibitors of the fibroblast growth factor receptor (FGFR) family of receptor tyrosine kinases. The compounds exhibit selective inhibition of FGFR over the closely related VEGFR2 receptor in cell lines and in vivo. The pharmacologic profile of these inhibitors was defined using a panel of human tumor cell lines characterized for specific mutations, amplifications, or translocations known to activate one of the four FGFR receptor isoforms. This pharmacology defines a profile for inhibitors that are likely to be of use in clinical settings in disease types where FGFR is shown to play an important role. Mol Cancer Ther; 10(9); 1542–52. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
28. Fragment-Based Drug Discovery Targeting Inhibitor of Apoptosis Proteins: Discovery of a Non-Alanine Lead Series with Dual Activity Against cIAP1 and XIAP
- Author
-
Darcey Miller, Aman Iqbal, Michael Reader, Gianni Chessari, Nicola E. Wilsher, Pamela A. Williams, Alison Jo-Anne Woolford, Christopher N. Johnson, George Ward, Vanessa Martins, Edward J. Lewis, David C. Rees, Ildiko Maria Buck, Philip J. Day, Sharna J. Rich, Marc Vitorino, Glyn Williams, James Edward Harvey Day, and Emiliano Tamanini
- Subjects
Models, Molecular ,Peptidomimetic ,Fragment-based lead discovery ,Antineoplastic Agents ,X-Linked Inhibitor of Apoptosis Protein ,Pharmacology ,Inhibitor of apoptosis ,Piperazines ,Inhibitor of Apoptosis Proteins ,Mice ,Drug Discovery ,Animals ,Humans ,Cell Proliferation ,Inhibitor of apoptosis domain ,Mice, Inbred BALB C ,Chemistry ,Drug discovery ,Computational Biology ,Xenograft Model Antitumor Assays ,Peptide Fragments ,XIAP ,High-Throughput Screening Assays ,Apoptosis ,Drug Design ,Cancer research ,Molecular Medicine ,Signal transduction - Abstract
Inhibitor of apoptosis proteins (IAPs) are important regulators of apoptosis and pro-survival signaling pathways whose deregulation is often associated with tumor genesis and tumor growth. IAPs have been proposed as targets for anticancer therapy, and a number of peptidomimetic IAP antagonists have entered clinical trials. Using our fragment-based screening approach, we identified nonpeptidic fragments binding with millimolar affinities to both cellular inhibitor of apoptosis protein 1 (cIAP1) and X-linked inhibitor of apoptosis protein (XIAP). Structure-based hit optimization together with an analysis of protein-ligand electrostatic potential complementarity allowed us to significantly increase binding affinity of the starting hits. Subsequent optimization gave a potent nonalanine IAP antagonist structurally distinct from all IAP antagonists previously reported. The lead compound had activity in cell-based assays and in a mouse xenograft efficacy model and represents a highly promising start point for further optimization.
- Published
- 2015
29. Abstract B161: Fragment-based discovery of a highly potent, orally bioavailable ERK1/2 inhibitor that modulates the phosphorylation and catalytic activity of ERK1/2
- Author
-
Nicola G. Wallis, Nick Palmer, Tom D. Heightman, Mike Reader, Sharna J. Rich, Hannah Braithwaite, Hugh Walton, James Edward Harvey Day, Valerio Berdini, Sandra Muench, Christopher W. Murray, Nicola E. Wilsher, Brent Graham, Neil Thompson, Harpreet K Saini, Charlotte Mary Griffiths-Jones, Caroline Richardson, Juan Castro, Vanessa Martins, Aurélie Courtin, Puja Pathuri, Marc O'Reilly, Charlotte East, David C. Rees, Joanne M. Munck, Alison Jo-Anne Woolford, David Norton, Lynsey Fazal, Ildiko Maria Buck, John Lyons, and Megan Cassidy
- Subjects
MAPK/ERK pathway ,Cancer Research ,Conformational change ,Oncology ,Cell culture ,Chemistry ,Cell growth ,Mutant ,Cancer research ,Phosphorylation ,Kinome ,V600E - Abstract
The RAS-RAF-MEK-ERK signalling cascade is activated through mutations in RAS or RAF in over 30% of cancers. The successful development of inhibitors of BRAF and MEK kinases has led to effective treatment particularly of melanomas whose tumor growth is driven by activating mutations in BRAF such as V600E. Despite these successes, resistance emerges after several months, leading to increased signaling through ERK1/2. This has prompted the development of direct inhibitors of ERK1/2, several of which are in early clinical trials. The majority of clinical ERK1/2 inhibitors are ATP competitive, blocking ERK1/2 catalytic phosphorylation of downstream substrates such as RSK, but do not modulate phosphorylation of ERK1/2 by MEK. Crystal structural studies performed by us and others on the pERK1/2 modulating inhibitor SCH772984 suggested that it induces a conformational change in the glycine-rich loop of ERK2, which leads to Tyr36 becoming tucked under the loop and creating a new binding pocket. We hypothesized that this binding mode might underlie the ability of SCH772984 to block the phosphorylation of ERK1/2, and initiated a fragment-based approach to develop novel, orally bioavailable inhibitors that elicit a similar conformational change and also modulate the phosphorylation of ERK1/2. Using screening methods including high-throughput X-ray crystallography and biophysical assays, we identified fragments binding to both the hinge and the inducible pocket of ERK2. Progressive rounds of structure-guided fragment optimization and growing led to an understanding of inhibitor structure determinants required to induce the conformational change in ERK2. These efforts, together with iterative optimization in a screening cascade including measurement of pRSK and pERK levels and antiproliferative activity in RAS and BRAF mutant cells, led to the discovery of a novel series of pERK modulating ERK1/2 inhibitors. The lead compound shows low nanomolar potency in biochemical ERK1/2 assays and an excellent kinome selectivity profile. In BRAF and RAS mutant cell lines, the lead shows low nanomolar cell proliferation IC50 values, while sparing cell lines not driven by the MAPK pathway. The lead exhibits robust antitumor activity upon oral dosing in a range of subcutaneous xenograft models including the mutant BRAF colorectal line Colo205, providing a promising basis for further optimization towards clinical pERK1/2 modulating ERK1/2 inhibitors. Citation Format: Tom D. Heightman, Valerio Berdini, Hannah Braithwaite, Ildiko Buck, Megan Cassidy, Juan Castro, Aurélie Courtin, James Day, Charlotte East, Lynsey Fazal, Brent Graham, Charlotte Griffiths-Jones, John Lyons, Vanessa Martins, Sandra Muench, Joanne Munck, David Norton, Marc O'Reilly, Nick Palmer, Puja Pathuri, Mike Reader, David Rees, Sharna Rich, Caroline Richardson, Harpreet Saini, Neil Thompson, Nicola Wallis, Hugh Walton, Nicola Wilsher, Alison Woolford, Chris Murray. Fragment-based discovery of a highly potent, orally bioavailable ERK1/2 inhibitor that modulates the phosphorylation and catalytic activity of ERK1/2 [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr B161.
- Published
- 2018
- Full Text
- View/download PDF
30. Abstract B154: Characterization of a novel ERK1/2 inhibitor, which modulates the phosphorylation and catalytic activity of ERK1/2
- Author
-
Luke Bevan, John Lyons, Caroline Richardson, Charlotte Mary Griffiths-Jones, Juan Castro, Lukas Stanczuk, Alpesh Shah, Hugh Walton, Neil T. Thompson, Harpreet K Saini, Nicola G. Wallis, Joanne M. Munck, Alison Jo-Anne Woolford, David Norton, Tom D. Heightman, Lynsey Fazal, Christopher J. Hindley, Christopher W. Murray, Hannah Braithwaite, Mike Reader, Vanessa Martins, Charlotte East, Puja Pathuri, Birikiti Kidane, Marc O'Reilly, David C. Rees, Aurélie Courtin, Megan Cassidy, Justyna Kucia-Tran, James Edward Harvey Day, Sandra Muench, Nicola E. Wilsher, Ildiko Maria Buck, Nick Palmer, Sharna J. Rich, Valerio Berdini, and Brent Graham
- Subjects
MAPK/ERK pathway ,Cancer Research ,Cell signaling ,biology ,Chemistry ,Receptor tyrosine kinase ,Substrate-level phosphorylation ,Oncology ,Downregulation and upregulation ,In vivo ,Cell culture ,biology.protein ,Cancer research ,Phosphorylation - Abstract
The MAPK pathway is commonly hyper-activated in human cancers due to the occurrence of oncogenic mutations in RAF, RAS and the upregulation of RTKs. The therapeutic potential of MAPK pathway inhibition has been demonstrated by the clinical efficacy of RAF and MEK1/2 (MEK) inhibitors in the treatment of BRAF-mutant melanoma. However, response to such agents is short-lived due to the onset of resistance mechanisms, which in the majority of cases result in the reactivation of ERK1/2 (ERK) signalling. Therefore, the direct targeting of ERK is an attractive therapeutic approach to overcoming the limitations of RAF or MEK inhibitors. Here, we describe a novel, potent, and selective ERK inhibitor, which inhibits both ERK catalytic activity and also the phosphorylation of ERK by MEK. Using fragment-based drug discovery we have developed a selective ERK inhibitor, which inhibits in vitro ERK catalytic activity with a low nM IC50 value. This lead compound has strong antiproliferative effects in a wide range of MAPK-activated cell lines, including the BRAF-mutant cell lines A375 (melanoma) and Colo205 (colorectal), the KRAS-mutant cell lines HCT116 (colorectal), Calu6 (lung) and Panc05.04 (pancreatic), and the NRAS-mutant cell line Ma-mel-27 (melanoma). The lead compound potently inhibits ERK cell signalling. The potent (nM) inhibition of RSK phosphorylation (a direct ERK substrate) was confirmed in A375 (BRAF-mutant melanoma) cells, using MSD analysis. In addition to inhibiting downstream ERK signalling, we demonstrated by ELISA and Western blotting that the lead compound confers a decrease in phospho-ERK levels in both BRAF-mutant and KRAS-mutant cell lines. We investigated the biochemical mechanism of the modulation of ERK phosphorylation in vitro and demonstrated that the compound prevents the phosphorylation of ERK by MEK (at key ERK activation loop residues, T202/Y204), without directly inhibiting MEK activity. The compound was profiled in a range of subcutaneous xenograft models including A375 (BRAF-mutant melanoma) and Calu-6 (KRAS-mutant lung). Once-daily oral dosing of the lead compound conferred significant antitumor activity in a range of in vivo efficacy studies. The compound potently inhibited the phosphorylation of downstream ERK substrates (including RSK) in tumor xenograft tissue. There was a clear relationship between in vivo compound concentrations and the modulation of ERK substrate phosphorylation. Furthermore, as was demonstrated in vitro, we confirmed that in addition to inhibiting ERK catalytic activity the compound potently inhibited the phosphorylation of ERK itself, in both KRAS and BRAF-mutant tumor xenografts. Here, we characterize a novel, highly potent, selective ERK inhibitor, which inhibits both ERK catalytic activity and also the upstream phosphorylation of ERK by MEK. These data support the further optimization of this series of compounds for clinical development. Citation Format: Joanne M. Munck, Valerio Berdini, Luke D. Bevan, Hannah Braithwaite, Ildiko M. Buck, Megan Cassidy, Juan Castro, Aurelie Courtin, James E. Day, Charlotte East, Lynsey Fazal, Brent Graham, Charlotte M. Griffiths-Jones, Tom D. Heightman, Chris J. Hindley, Birikiti Kidane, Justyna Kucia-Tran, John F. Lyons, Vanessa Martins, Sandra Muench, Chris W. Murray, David Norton, Marc O'Reilly, Nick Palmer, Puja Pathuri, Mike Reader, David C. Rees, Sharna J. Rich, Caroline J. Richardson, Harpreet K. Saini, Alpesh Shah, Lukas Stanczuk, Neil T. Thompson, Hugh Walton, Nicola E. Wilsher, Alison J. Woolford, Nicola G. Wallis. Characterization of a novel ERK1/2 inhibitor, which modulates the phosphorylation and catalytic activity of ERK1/2 [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr B154.
- Published
- 2018
- Full Text
- View/download PDF
31. Discovery of (2,4-dihydroxy-5-isopropylphenyl)-[5-(4-methylpiperazin-1-ylmethyl)-1,3-dihydroisoindol-2-yl]methanone (AT13387), a novel inhibitor of the molecular chaperone Hsp90 by fragment based drug design
- Author
-
Maria Grazia Carr, Jonathan Lewis, Gary Trewartha, Ruth Feltell, Brian John Williams, Martyn Frederickson, Lina Parra, Andrew James Woodhead, Alison Jo-Anne Woolford, Joseph E. Coyle, Christopher W. Murray, Lynsey Fazal, Theresa Rachael Phillips, Donna-Michelle Smith, Rachel McMenamin, Brent Graham, Eva Figueroa, Sharna J. Rich, Gianni Chessari, M. Alistair O’Brien, Miles Congreve, Sahil Patel, Jose Cosme, David C. Rees, Robert Downham, Philip J. Day, Hayley Angove, and Mladen Vinkovic
- Subjects
Drug ,Models, Molecular ,media_common.quotation_subject ,Transplantation, Heterologous ,Molecular Conformation ,Mice, Nude ,HSP90 Heat-Shock Proteins ,Antineoplastic Agents ,Pharmacology ,Isoindoles ,Crystallography, X-Ray ,Ligands ,Cell Line ,Mice ,Structure-Activity Relationship ,Drug Stability ,Heat shock protein ,Drug Discovery ,Structure–activity relationship ,Animals ,Humans ,Tissue Distribution ,media_common ,Mice, Inbred BALB C ,biology ,Chemistry ,Drug discovery ,HCT116 Cells ,Hsp90 ,Combinatorial chemistry ,Transplantation ,Solubility ,Chaperone (protein) ,Drug Design ,Benzamides ,biology.protein ,Molecular Medicine ,Female ,Drug Screening Assays, Antitumor ,Neoplasm Transplantation - Abstract
Inhibitors of the molecular chaperone heat shock protein 90 (Hsp90) are currently generating significant interest in clinical development as potential treatments for cancer. In a preceding publication (DOI: 10.1021/jm100059d ) we describe Astex's approach to screening fragments against Hsp90 and the subsequent optimization of two hits into leads with inhibitory activities in the low nanomolar range. This paper describes the structure guided optimization of the 2,4-dihydroxybenzamide lead molecule 1 and details some of the drug discovery strategies employed in the identification of AT13387 (35), which has progressed through preclinical development and is currently being tested in man.
- Published
- 2010
32. Identification of novel allosteric inhibitors through fragment-based drug discovery and X-ray crystallography
- Author
-
O'Brien, P.A. Williams, G. Chessari, M.G. Carr, S. Hiscock, P. Pathuri, Joseph E. Coyle, M. Vinkovic, T. Sambrook, C. Richardson, Anne Cleasby, J.R. Yon, M. Congreve, S. Saalau-Bethell, H. Jhoti, V. Lock, A.J. Woodhead, Christopher William Murray, B. Graham, V Berdini, and Sharna J. Rich
- Subjects
Inorganic Chemistry ,Structural Biology ,Chemistry ,Stereochemistry ,Fragment-based lead discovery ,X-ray crystallography ,Allosteric regulation ,General Materials Science ,Identification (biology) ,Physical and Theoretical Chemistry ,Condensed Matter Physics ,Biochemistry - Published
- 2015
- Full Text
- View/download PDF
33. 380 Induction of apoptosis with a novel dual cIAP1/XIAP antagonist in models of melanoma
- Author
-
C.N. Johnson, Neil T. Thompson, Jon Lewis, G. Ward, G. Chessari, and Sharna J. Rich
- Subjects
Cancer Research ,Oncology ,business.industry ,Apoptosis ,Melanoma ,Antagonist ,Cancer research ,Medicine ,DUAL (cognitive architecture) ,business ,medicine.disease ,XIAP - Published
- 2014
- Full Text
- View/download PDF
34. Abstract 4748: Discovery of JNJ-42756493, a potent fibroblast growth factor receptor (FGFR) inhibitor using a fragment based approach
- Author
-
Sharna J. Rich, Jorge Vialard, Eleonora Jovcheva, Delphine Yvonne Raymonde Lardeau, David R. Newell, Virginie Poncelet, Helene Colombel, Anne Cleasby, Christopher William Murray, Christophe Denis Adelinet, Tinne Verhulst, Elisabeth Thérèse Jeanne Pasquier, Martin Page, Steve Mcclue, Matthew S Squires, Yannick Ligny, Jean Fernand Armand Lacrampe, Peter King, Yvan Simonnet, Timothy Perera, Laurence Mevellec, Patrick René Angibaud, Olivier Alexis Georges Querolle, Pilatte Isabelle Noelle Consta, Marc Willems, Gordon Saxty, Lieven Meerpoel, Werner Constant Johan Embrechts, Eddy Jean Edgard Freyne, Pascal Bonnet, Bruno Roux, Virginie Tronel, Rhalid Akkari, Valerio Berdini, Steven John Woodhead, Elodie Sement, Berthold Wroblowski, David C. Rees, Xavier Bourdrez, Imre Christian Francis Csoka, Marine Bourgeois, Ronaldus Arnodus Hendrika Joseph Gilissen, and Alexandra Papanikos
- Subjects
Cancer Research ,Oncology ,Fibroblast growth factor receptor ,Drug discovery ,Fibroblast growth factor receptor 1 ,Human gastric carcinoma ,Biology ,Kinase inhibition ,Receptor ,Fibroblast growth factor ,Molecular biology - Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFR1 through 4) regulate a variety of key cellular processes, including proliferation, migration, survival, and differentiationa. Aberrant activation of FGF/FGFR is strongly implicated in oncogenic signalling in many tumor types. This has stimulated the development of a number of FGFR inhibitors, with diverse kinase inhibition and pharmacological profiles that are currently being evaluated in clinical studies. We conducted a fragment screening campaign and this resulted in identification of a 6-aminoquinoxalinyl fragment with a binding affinity in the micromolar range. Structure-guided medicinal chemistry led to the identification of a novel quinoxaline-based chemical series with nanomolar affinity for FGFR1, 2, 3, and 4, activity in cells, and selectivity with respect to VEGFR-2. Further optimisation resulted in the generation of JNJ-42756493, a compound with favourable drug-like properties that demonstrated strong anti-tumoral activity in a FGFR2-dependent SNU-16 human gastric carcinoma xenograft model. This report represents the first disclosure of the structure-activity relationships as well as the chemical synthesis pathway of the JNJ-42756493 series and illustrates how a fragment-based drug discovery approach has been efficiently used to discover FGFR1-4 inhibitors with nanomolar affinity. aTurner, N. and Grose, R. Nat. Rev. Cancer, 2010, 10, 116-129. Citation Format: Patrick R. Angibaud, Laurence Mevellec, Gordon Saxty, Christophe Adelinet, Rhalid Akkari, Valerio Berdini, Pascal Bonnet, Marine Bourgeois, Xavier Bourdrez, Anne Cleasby, Helene Colombel, Imre Csoka, Werner Embrechts, Eddy Freyne, Ronaldus Gilissen, Eleonora Jovcheva, Peter King, Jean Lacrampe, Delphine Lardeau, Yannick Ligny, Steve Mcclue, Lieven Meerpoel, David R. Newell, Martin Page, Alexandra Papanikos, Elisabeth Pasquier, Isabelle Pilatte, Virginie Poncelet, Olivier Querolle, David C. Rees, Sharna Rich, Bruno Roux, Elodie Sement, Yvan Simonnet, Matthew Squires, Virginie Tronel, Tinne Verhulst, Jorge Vialard, Marc Willems, Steven J. Woodhead, Berthold Wroblowski, Christopher W. Murray, Timothy Perera. Discovery of JNJ-42756493, a potent fibroblast growth factor receptor (FGFR) inhibitor using a fragment based approach. [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 4748. doi:10.1158/1538-7445.AM2014-4748
- Published
- 2014
- Full Text
- View/download PDF
35. Discovery of novel allosteric inhibitors of HCV NS3/4a enzyme via structure-based drug design
- Author
-
H. Jhoti, J.R. Yon, A.J. Woodhead, M.G. Carr, C. Richardson, Neil T. Thompson, P. Pathuri, R. McMenamin, Joseph E. Coyle, S. Saalau-Bethell, M. Verdonk, Sharna J. Rich, Christopher William Murray, C.C.F. Hamlett, David C. Rees, B. Graham, P.A. Williams, G. Chessari, S. Hiscock, M. Vinkovic, M. Frederickson, M. Reader, and F.P. Holding
- Subjects
Structural Biology ,media_common.quotation_subject ,Allosteric regulation ,Art history ,Structure based ,Art ,media_common - Abstract
NS3/4a enzyme via structure-based drug design Puja Pathuri, Susanne M. Saalau-Bethell, Andrew J. Woodhead, Maria G. Carr, Gianni Chessari, Joseph Coyle, a Martyn Frederickson, Brent Graham, Chris Hamlett, Steven D. Hiscock, Finn P. Holding, Harren Jhoti, Rachel McMenamin, Chris W. Murray, Mike Reader, David C. Rees, Sharna J. Rich, Caroline J. Richardson, Neil Thompson, Marcel L. Verdonk, Mladen Vinkovic, Pamela A. Williams, Jeff Yon Astex Pharmaceuticals, 436 Cambridge Science Park, Cambridge, CB4 0QA, UK E-mail: Puja.Pathuri@astx.com
- Published
- 2012
- Full Text
- View/download PDF
36. 871 FRAGMENT BASED DISCOVERY AND GENOTYPIC PROFILING OF A SERIES OF DIRECT ACTING ANTIVIRAL AGENTS WITH A NOVEL MODE OF ACTION
- Author
-
S. Saalau-Bethell, Neil T. Thompson, A.J. Woodhead, H. Willems, Joseph E. Coyle, M. Vinkovic, S. Hiscock, H. Jhotti, M. Verdonk, F.P. Holding, P.A. Williams, G. Chessari, C. Richardson, C. Murray, Sharna J. Rich, and Brent Graham
- Subjects
Hepatology ,Genotype ,Profiling (information science) ,Computational biology ,Pharmacology ,Biology ,Mode of action ,Direct acting - Published
- 2012
- Full Text
- View/download PDF
37. 878 PRE-CLINICAL CHARACTERISATION OF A NOVEL DIRECT ACTING ANTIVIRAL AGENT WITH A NEW MODE OF ACTION
- Author
-
A. Millemaggi, A.J. Woodhead, H. Willems, M. Carr, M. Reader, C.C.F. Hamlett, S. Saalau-Bethell, Brent Graham, L. Fazal, Nicola E. Wilsher, E. Chiarparin, Vanessa Martins, Jon Lewis, M. Hodder, C. Murray, Sharna J. Rich, M. Sanders, David C. Rees, S. Hiscock, Neil T. Thompson, D. Norton, and P. Pathuri
- Subjects
Hepatology ,Chemistry ,Mode of action ,Neuroscience ,Direct acting - Published
- 2012
- Full Text
- View/download PDF
38. Abstract 1361: Fragment based drug discovery of selective inhibitors of Fibroblast Growth Factor Receptor (FGFR)
- Author
-
David C. Rees, Timothy Perera, E Sement, Martin Page, Yannick Ligny, Christopher William Murray, P Benderitter, Rhalid Akkari, Alexandra Papanikos, Andrew Madin, Laurence Anne Mevellec, Janine Arts, Berthold Wroblowski, Y Simmonet, Steve Mcclue, M. Squires, Neil T. Thompson, Ronaldus Arnodus Hendrika Joseph Gilissen, V Berdini, Gordon Saxty, Sharna J. Rich, Pascal Bonnet, Anne Cleasby, H Newell, S. Saalau-Bethell, V. Tronel, Freyne Eddy Jean Edgard, Werner Constant Johan Embrechts, Peter King, Olivier Querolle, Patrick Angibaud, Marc Willems, Jean Fernand Armand Lacrampe, and George Ward
- Subjects
Cancer Research ,Kinase ,Molecular pathology ,Chemistry ,Fibroblast growth factor receptor 1 ,Fragment-based lead discovery ,Cancer ,medicine.disease ,Fibroblast growth factor ,Oncology ,Biochemistry ,Fibroblast growth factor receptor ,medicine ,Cancer research ,Tyrosine kinase - Abstract
Recent data in a number of tumour types has implicated Fibroblast Growth Factor (FGF) and Fibroblast Growth Factor receptor (FGFR) signalling as being key to the molecular pathology of cancer. A fragment screening campaign was conducted against the tyrosine kinase domain of FGFR1 to detect low molecular weight compounds that bound to the hinge region of the kinase. The screening produced several fragment inhibitors (molecular weight The poster will focus on the description of previously undescribed compounds bearing an imidazo[1,2-a]pyridine core scaffold where selectivity versus other protein kinases, for example FLT3, is obtained using the X-ray crystal structure and structure-based design. In summary we will illustrate how X-ray crystallography and fragment-based drug design (FBDD) can be used to discover compounds with activity in an FGFR driven xenograft model when dosed by the oral route. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1361. doi:10.1158/1538-7445.AM2011-1361
- Published
- 2011
- Full Text
- View/download PDF
39. Abstract 3626: Development of inhibitors of the fibroblast growth factor receptor (FGFR) kinase using a fragment based approach
- Author
-
Mike A. Batey, Isobel Harada, Ruth Feltell, Gordon Saxty, Neil T. Thompson, George Ward, Edward J. Lewis, Yan Zhao, Timothy Perera, Julie Irving, David R. Newell, Christopher William Murray, Peter H. King, Lynsey Fazal, Ron Gilissen, Sharna J. Rich, Matthew S Squires, and Patrik Angibaud
- Subjects
Cancer Research ,biology ,Kinase ,Cell growth ,FGFR Inhibition ,Fibroblast growth factor ,Receptor tyrosine kinase ,Oncology ,Biochemistry ,Fibroblast growth factor receptor ,Cancer research ,biology.protein ,Protein kinase A ,Protein kinase B - Abstract
Recent data in a number of tumour types has implicated Fibroblast Growth Factor (FGF) and Fibroblast Growth Factor receptor (FGFR) signalling as being key to the molecular pathology of cancer. FGFR is a receptor tyrosine kinase which activates the extracellular signal-regulated kinase / mitogen-activated protein kinase and the protein kinase B / Akt pathways which promote cell growth and survival. Amplification, over-expression or activating mutations of fibroblast growth factor receptors have been associated with bladder tumours, multiple myeloma, hormone-refractory prostate cancer and breast cancer. Multiple lead series of FGFR inhibitors were developed using Astex's fragment based medicinal chemistry approach, Pyramid™, linked to high throughput X-ray Crystallography. We describe here the characterisation of some examples of these lead molecules. In particular we detail the pharmacological profile of a compound from one of these lead series that demonstrated activity against FGFR 1-4 with an IC50 10 fold lower in cell lines lacking FGFR expression. We demonstrate inhibition of FGFR 2 and 3 phosphorylation in gastric and multiple myeloma cell lines respectively with associated inhibition of downstream signalling pathways. This lead molecule has an excellent pharmacokinetic profile and high oral bioavailibility in mice and rats. In xenograft models in mice where aberrant FGF signalling underlies tumour pathology, tumour growth inhibition is observed at doses of 100mg/kg /day orally for 21 days. This xenograft efficacy was observed in several models, with significantly lower activity in models where aberrant FGF signalling is not involved in tumour pathology. This suggests that the mechanism of action is consistent with FGFR inhibition. The pharmacological profile in these models is also distinct from other broader spectrum receptor tyrosine kinase inhibitors. The pre-clinical data shown here suggests that such compounds warrant further investigation pre-clinically and may benefit patients whose disease is driven by FGFR activity. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3626.
- Published
- 2010
- Full Text
- View/download PDF
40. Abstract 5778: Fragment-based drug discovery of selective inhibitors of fibroblast growth factor receptor (FGFr)
- Author
-
C Griffiths Jones, Brent Graham, Timothy Perera, Maria Grazia Carr, Rajdeep Kaur Nijjar, Andrew Madin, E Vickerstaff, S. Saalau-Bethell, Anne Cleasby, V Berdini, Darcey Miller, Edward J. Lewis, Joseph E. Coyle, Gordon Saxty, George Ward, Andrew Pike, Michael Alistair O'brien, Christopher William Murray, H Newell, Sharna J. Rich, Patrick Angibaud, and M. Squires
- Subjects
Cancer Research ,chemistry.chemical_compound ,Oncology ,Biochemistry ,Molecular pathology ,Fibroblast growth factor receptor ,Chemistry ,Kinase ,Drug discovery ,Fragment-based lead discovery ,Target protein ,Fibroblast growth factor ,Lead compound - Abstract
Recent data in a number of tumour types has implicated Fibroblast Growth Factor (FGF) and Fibroblast Growth Factor receptor (FGFr) signalling as being key to the molecular pathology of cancer. This poster will describe fragment based drug discovery using biophysical screening to identify initial fragments. Subsequently, in the fragments-to-leads stage a detailed structural understanding of the binding interactions between the fragment and its target protein utilised X-ray crystallography and NMR. Starting with different fragments allows several lead series to be identified, often by synthesizing only small numbers of compounds. A fragment screening campaign was conducted against the FGFr-1 to detect very low molecular weight compounds that bound to the hinge region of the kinase. The screening produced several fragment molecules (Molecular Weight This poster represents first disclosure of the structure of the lead series and illustrates how a fragment-based drug discovery approach can be efficiently used to discover compounds advanced nanomolar compounds with oral bioavailability. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5778.
- Published
- 2010
- Full Text
- View/download PDF
41. Fragment-BasedDrug Discovery Targeting Inhibitorof Apoptosis Proteins: Discovery of a Non-Alanine Lead Series withDual Activity Against cIAP1 and XIAP.
- Author
-
Gianni Chessari, Ildiko M. Buck, James E. H. Day, PhilipJ. Day, Aman Iqbal, Christopher N. Johnson, Edward J. Lewis, Vanessa Martins, Darcey Miller, Michael Reader, David C. Rees, Sharna J. Rich, Emiliano Tamanini, Marc Vitorino, George A. Ward, Pamela A. Williams, Glyn Williams, Nicola E. Wilsher, and AlisonJ.-A. Woolford
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.