Network


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

Hotspot


Dive into the research topics where Paul Lyne is active.

Publication


Featured researches published by Paul Lyne.


Drug Discovery Today | 2002

Structure-based virtual screening: an overview.

Paul Lyne

Enormous advances in genomics have resulted in a large increase in the number of potential therapeutic targets that are available for investigation. This growth in potential targets has increased the demand for reliable target validation, as well as technologies that can identify rapidly several quality lead candidates. Virtual screening, and in particular receptor-based virtual screening, has emerged as a reliable, inexpensive method for identifying leads. Although still an evolving method, advances in computational techniques have enabled virtual screening to have a positive impact on the discovery process. Here, the current strengths and weaknesses of the technology are discussed, and emphasis is placed on aspects of the work-flow of a virtual screening campaign, from preparation through to post-screening analysis.


Journal of Chemical Information and Modeling | 2006

On Evaluating Molecular-Docking Methods for Pose Prediction and Enrichment Factors

Hongming Chen; Paul Lyne; Fabrizio Giordanetto; Timothy Lovell; Jin Li

Four of the most well-known, commercially available docking programs, FlexX, GOLD, GLIDE, and ICM, have been examined for their ligand-docking and virtual-screening capabilities. The relative performance of the programs in reproducing the native ligand conformation from starting SMILES strings for 164 high-resolution protein-ligand complexes is presented and compared. Applying only the native scoring functions, the latest versions of these four docking programs were also used to conduct virtual screening for 12 protein targets of therapeutic interest, involving both publicly available structures and AstraZeneca in-house structures. The capability of the four programs to correctly rank-order target-specific active compounds over alternative binders and nonbinders (decoys plus randomly selected compounds) and thereby enrich a small subset of a screening library is compared. Enrichments from the virtual-screening experiments are contrasted with those obtained with alternative 3D shape-matching and 2D similarity database-search methods.


Blood | 2014

AZD1208, a potent and selective pan-Pim kinase inhibitor, demonstrates efficacy in preclinical models of acute myeloid leukemia

Erika K. Keeton; Kristen McEachern; Keith Dillman; Sangeetha Palakurthi; Yichen Cao; Michael Grondine; Surinder Kaur; Suping Wang; Yuching Chen; Allan Wu; Minhui Shen; Francis D. Gibbons; Michelle Lamb; Xiaolan Zheng; Richard Stone; Daniel J. DeAngelo; Leonidas C. Platanias; Les A. Dakin; Huawei Chen; Paul Lyne; Dennis Huszar

Upregulation of Pim kinases is observed in several types of leukemias and lymphomas. Pim-1, -2, and -3 promote cell proliferation and survival downstream of cytokine and growth factor signaling pathways. AZD1208 is a potent, highly selective, and orally available Pim kinase inhibitor that effectively inhibits all three isoforms at <5 nM or <150 nM in enzyme and cell assays, respectively. AZD1208 inhibited the growth of 5 of 14 acute myeloid leukemia (AML) cell lines tested, and sensitivity correlates with Pim-1 expression and STAT5 activation. AZD1208 causes cell cycle arrest and apoptosis in MOLM-16 cells, accompanied by a dose-dependent reduction in phosphorylation of Bcl-2 antagonist of cell death, 4EBP1, p70S6K, and S6, as well as increases in cleaved caspase 3 and p27. Inhibition of p4EBP1 and p-p70S6K and suppression of translation are the most representative effects of Pim inhibition in sensitive AML cell lines. AZD1208 inhibits the growth of MOLM-16 and KG-1a xenograft tumors in vivo with a clear pharmacodynamic-pharmacokinetic relationship. AZD1208 also potently inhibits colony growth and Pim signaling substrates in primary AML cells from bone marrow that are Flt3 wild-type or Flt3 internal tandem duplication mutant. These results underscore the therapeutic potential of Pim kinase inhibition for the treatment of AML.


Journal of Medicinal Chemistry | 2008

Identification of 4-aminopyrazolylpyrimidines as potent inhibitors of Trk kinases.

Tao Wang; Michelle L. Lamb; David Scott; Haixia Wang; Michael Howard Block; Paul Lyne; John W. Lee; Audrey Davies; Hai-Jun Zhang; Yanyi Zhu; Fei Gu; Yongxin Han; Bin Wang; Peter Mohr; Robert J. Kaus; John Anthony Josey; Ethan Hoffmann; Ken Thress; Terry MacIntyre; Haiyun Wang; Charles Omer; Dingwei Yu

The design, synthesis and biological evaluation of a series of 4-aminopyrazolylpyrimidines as potent Trk kinase inhibitors is reported. High-throughput screening identified a promising hit in the 4-aminopyrazolylpyrimidine chemotype. Initial optimization of the series led to more potent Trk inhibitors. Further optimization using two strategies resulted in significant improvement of physical properties and led to the discovery of 10z (AZ-23), a potent, orally bioavailable Trk A/B inhibitor. The compound offers the potential to test the hypothesis that modulation of Trk activity will be of benefit in the treatment of cancer and other indications in vivo.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of novel benzylidene-1,3-thiazolidine-2,4-diones as potent and selective inhibitors of the PIM-1, PIM-2, and PIM-3 protein kinases.

Les A. Dakin; Michael Howard Block; Huawei Chen; Erin Code; James E. Dowling; Xiaomei Feng; Andrew D. Ferguson; Isabelle Green; Alexander Hird; Tina Howard; Erika K. Keeton; Michelle Lamb; Paul Lyne; Hannah Pollard; Jon Read; Allan Wu; Tao Zhang; Xiaolan Zheng

Novel substituted benzylidene-1,3-thiazolidine-2,4-diones (TZDs) have been identified as potent and highly selective inhibitors of the PIM kinases. The synthesis and SAR of these compounds are described, along with X-ray crystallographic, anti-proliferative, and selectivity data.


Journal of Medicinal Chemistry | 2012

Discovery of Checkpoint Kinase Inhibitor (S)-5-(3-Fluorophenyl)-N-(piperidin-3-yl)-3-ureidothiophene-2-carboxamide (AZD7762) by Structure-Based Design and Optimization of Thiophenecarboxamide Ureas.

Oza; Susan Ashwell; Lynsie Almeida; Patrick Brassil; Jason Breed; Chun Deng; Thomas Gero; Michael Grondine; C Horn; Stephanos Ioannidis; D Liu; Paul Lyne; Nicholas John Newcombe; Martin Pass; Jon Read; S Ready; S Rowsell; Mei Su; Dorin Toader; Melissa Vasbinder; Dingwei Yu; Yan Yu; Y Xue; S Zabludoff; James W. Janetka

Checkpoint kinases CHK1 and CHK2 are activated in response to DNA damage that results in cell cycle arrest, allowing sufficient time for DNA repair. Agents that lead to abrogation of such checkpoints have potential to increase the efficacy of such compounds as chemo- and radiotherapies. Thiophenecarboxamide ureas (TCUs) were identified as inhibitors of CHK1 by high throughput screening. A structure-based approach is described using crystal structures of JNK1 and CHK1 in complex with 1 and 2 and of the CHK1-3b complex. The ribose binding pocket of CHK1 was targeted to generate inhibitors with excellent cellular potency and selectivity over CDK1and IKKβ, key features lacking from the initial compounds. Optimization of 3b resulted in the identification of a regioisomeric 3-TCU lead 12a. Optimization of 12a led to the discovery of the clinical candidate 4 (AZD7762), which strongly potentiates the efficacy of a variety of DNA-damaging agents in preclinical models.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery of a novel class of 2-ureido thiophene carboxamide checkpoint kinase inhibitors.

James W. Janetka; Lynsie Almeida; Susan Ashwell; Patrick Brassil; Kevin Daly; Chun Deng; Thomas Gero; Roberta Glynn; Candice Horn; Stephanos Ioannidis; Paul Lyne; Nicholas John Newcombe; Vibha Oza; Martin Pass; Stephanie Springer; Mei Su; Dorin Toader; Melissa Vasbinder; Dingwei Yu; Yan Yu; Sonya Zabludoff

Checkpoint kinase-1 (Chk1, CHEK1) is a Ser/Thr protein kinase that mediates the cellular response to DNA-damage. A novel class of 2-ureido thiophene carboxamide urea (TCU) Chk1 inhibitors is described. Inhibitors in this chemotype were optimized for cellular potency and selectivity over Cdk1.


Molecular Cancer Therapeutics | 2016

AZD5153: a novel bivalent BET bromodomain inhibitor highly active against hematologic malignancies

Garrett W. Rhyasen; Maureen Hattersley; Yi Yao; Austin Dulak; Wenxian Wang; Philip Petteruti; Ian L. Dale; Scott Boiko; Tony Cheung; Jingwen Zhang; Shenghua Wen; Lillian Castriotta; Deborah Lawson; Mike Collins; Larry Bao; Miika Ahdesmaki; Graeme Walker; Greg O'Connor; Tammie C. Yeh; Alfred A. Rabow; Jonathan R. Dry; Corinne Reimer; Paul Lyne; Gordon B. Mills; Stephen Fawell; Michael J. Waring; Michael Zinda; Edwin Clark; Huawei Chen

The bromodomain and extraterminal (BET) protein BRD4 regulates gene expression via recruitment of transcriptional regulatory complexes to acetylated chromatin. Pharmacological targeting of BRD4 bromodomains by small molecule inhibitors has proven to be an effective means to disrupt aberrant transcriptional programs critical for tumor growth and/or survival. Herein, we report AZD5153, a potent, selective, and orally available BET/BRD4 bromodomain inhibitor possessing a bivalent binding mode. Unlike previously described monovalent inhibitors, AZD5153 ligates two bromodomains in BRD4 simultaneously. The enhanced avidity afforded through bivalent binding translates into increased cellular and antitumor activity in preclinical hematologic tumor models. In vivo administration of AZD5153 led to tumor stasis or regression in multiple xenograft models of acute myeloid leukemia, multiple myeloma, and diffuse large B-cell lymphoma. The relationship between AZD5153 exposure and efficacy suggests that prolonged BRD4 target coverage is a primary efficacy driver. AZD5153 treatment markedly affects transcriptional programs of MYC, E2F, and mTOR. Of note, mTOR pathway modulation is associated with cell line sensitivity to AZD5153. Transcriptional modulation of MYC and HEXIM1 was confirmed in AZD5153-treated human whole blood, thus supporting their use as clinical pharmacodynamic biomarkers. This study establishes AZD5153 as a highly potent, orally available BET/BRD4 inhibitor and provides a rationale for clinical development in hematologic malignancies. Mol Cancer Ther; 15(11); 2563–74. ©2016 AACR.


Journal of Medicinal Chemistry | 2013

Discovery and Optimization of a Novel Series of Potent Mutant B-Raf V600E Selective Kinase Inhibitors.

Melissa Vasbinder; Brian Aquila; Martin Augustin; Huawei Chen; Tony Cheung; Donald J. Cook; Lisa Drew; Benjamin P. Fauber; Steve Glossop; Michael Grondine; Edward J. Hennessy; Jeffrey W. Johannes; Stephen Lee; Paul Lyne; Mario Mörtl; Charles Omer; Sangeetha Palakurthi; Timothy Pontz; Jon Read; Li Sha; Minhui Shen; Stefan Steinbacher; Haixia Wang; Allan Wu; Minwei Ye

B-Raf represents an attractive target for anticancer therapy and the development of small molecule B-Raf inhibitors has delivered new therapies for metastatic melanoma patients. We have discovered a novel class of small molecules that inhibit mutant B-Raf(V600E) kinase activity both in vitro and in vivo. Investigations into the structure-activity relationships of the series are presented along with efforts to improve upon the cellular potency, solubility, and pharmacokinetic profile. Compounds selectively inhibited B-Raf(V600E) in vitro and showed preferential antiproliferative activity in mutant B-Raf(V600E) cell lines and exhibited selectivity in a kinase panel against other kinases. Examples from this series inhibit growth of a B-Raf(V600E) A375 xenograft in vivo at a well-tolerated dose. In addition, aminoquinazolines described herein were shown to display pERK elevation in nonmutant B-Raf cell lines in vitro.


ACS Medicinal Chemistry Letters | 2013

Structure and Property Based Design of Pyrazolo[1,5-a]pyrimidine Inhibitors of CK2 Kinase with Activity in Vivo.

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.

Collaboration


Dive into the Paul Lyne's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge