Network


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

Hotspot


Dive into the research topics where Mark Ledeboer is active.

Publication


Featured researches published by Mark Ledeboer.


Journal of Medicinal Chemistry | 2014

Discovery of a Novel, First-in-Class, Orally Bioavailable Azaindole Inhibitor (VX-787) of Influenza PB2.

M.P Clark; Mark Ledeboer; I Davies; R.A Byrn; S.M Jones; Emanuele Perola; A Tsai; Marc Jacobs; K Nti-Addae; Upul K. Bandarage; M.J Boyd; R.S Bethiel; John J. Court; H Deng; John P. Duffy; W.A Dorsch; Luc J. Farmer; Huai Gao; Wenxin Gu; K Jackson; D.H Jacobs; J.M Kennedy; Brian Ledford; J Liang; Francois Maltais; Mark A. Murcko; Tiansheng Wang; M.W Wannamaker; H.B Bennett; Joshua R. Leeman

In our effort to develop agents for the treatment of influenza, a phenotypic screening approach utilizing a cell protection assay identified a series of azaindole based inhibitors of the cap-snatching function of the PB2 subunit of the influenza A viral polymerase complex. Using a bDNA viral replication assay (Wagaman, P. C., Leong, M. A., and Simmen, K. A. Development of a novel influenza A antiviral assay. J. Virol. Methods 2002, 105, 105-114) in cells as a direct measure of antiviral activity, we discovered a set of cyclohexyl carboxylic acid analogues, highlighted by VX-787 (2). Compound 2 shows strong potency versus multiple influenza A strains, including pandemic 2009 H1N1 and avian H5N1 flu strains, and shows an efficacy profile in a mouse influenza model even when treatment was administered 48 h after infection. Compound 2 represents a first-in-class, orally bioavailable, novel compound that offers potential for the treatment of both pandemic and seasonal influenza and has a distinct advantage over the current standard of care treatments including potency, efficacy, and extended treatment window.


Antimicrobial Agents and Chemotherapy | 2015

Preclinical Activity of VX-787, a First-in-Class, Orally Bioavailable Inhibitor of the Influenza Virus Polymerase PB2 Subunit

Randal Byrn; Steven M. Jones; Hamilton Bennett; Chris M Bral; Michael P. Clark; Marc Jacobs; Ann D. Kwong; Mark Ledeboer; Joshua R. Leeman; Colleen F. McNeil; Mark A. Murcko; Azin Nezami; Emanuele Perola; Rene Rijnbrand; Kumkum Saxena; Alice W. Tsai; Yi Zhou; Paul S. Charifson

ABSTRACT VX-787 is a novel inhibitor of influenza virus replication that blocks the PB2 cap-snatching activity of the influenza viral polymerase complex. Viral genetics and X-ray crystallography studies provide support for the idea that VX-787 occupies the 7-methyl GTP (m7GTP) cap-binding site of PB2. VX-787 binds the cap-binding domain of the PB2 subunit with a KD (dissociation constant) of 24 nM as determined by isothermal titration calorimetry (ITC). The cell-based EC50 (the concentration of compound that ensures 50% cell viability of an uninfected control) for VX-787 is 1.6 nM in a cytopathic effect (CPE) assay, with a similar EC50 in a viral RNA replication assay. VX-787 is active against a diverse panel of influenza A virus strains, including H1N1pdm09 and H5N1 strains, as well as strains with reduced susceptibility to neuraminidase inhibitors (NAIs). VX-787 was highly efficacious in both prophylaxis and treatment models of mouse influenza and was superior to the neuraminidase inhibitor, oseltamivir, including in delayed-start-to-treat experiments, with 100% survival at up to 96 h postinfection and partial survival in groups where the initiation of therapy was delayed up to 120 h postinfection. At different doses, VX-787 showed a 1-log to >5-log reduction in viral load (relative to vehicle controls) in mouse lungs. Overall, these favorable findings validate the PB2 subunit of the viral polymerase as a drug target for influenza therapy and support the continued development of VX-787 as a novel antiviral agent for the treatment of influenza infection.


Bioorganic & Medicinal Chemistry Letters | 2010

A novel chemotype of kinase inhibitors: Discovery of 3,4-ring fused 7-azaindoles and deazapurines as potent JAK2 inhibitors.

Tiansheng Wang; Mark Ledeboer; John P. Duffy; Albert Pierce; Harmon Zuccola; Eric Block; Dina Shlyakter; James K. Hogan; Youssef L. Bennani

Pictet-Spengler condensation of aldehydes or alpha-keto-esters with 4-(2-anilinophenyl)-7-azaindole (11) or deazapurine (12) gave high yields of the 3,4-fused cyclic compounds. SAR studies, by varying the substituted benzaldehyde components, lead to the discovery of a series of potent JAK2 kinase inhibitors.


Journal of Medicinal Chemistry | 2009

Janus Kinase 2 Inhibitors. Synthesis and Characterization of a Novel Polycyclic Azaindole

Tiansheng Wang; John P. Duffy; Jian Wang; Summer Halas; Francesco G. Salituro; Albert Pierce; Harmon Zuccola; James R. Black; James K. Hogan; Scott Jepson; Dina Shlyakter; Sudipta Mahajan; Yong Gu; Thomas Hoock; Mark Wood; Brinley Furey; J. Daniel Frantz; Lisa M. Dauffenbach; Ursula A. Germann; Bin Fan; Mark Namchuk; Youssef L. Bennani; Mark Ledeboer

The synthesis and characterization of a novel polycyclic azaindole based derivative is disclosed, and its binding to JAK2 is described. The compound is further evaluated for its ability to block the EPO/JAK2 signaling cascade in vitro and in vivo.


Journal of Medicinal Chemistry | 2015

Discovery of VX-509 (Decernotinib): A Potent and Selective Janus Kinase 3 Inhibitor for the Treatment of Autoimmune Diseases.

L.J Farmer; Mark Ledeboer; T Hoock; M.J Arnost; R.S Bethiel; Youssef L. Bennani; J.J Black; C.L Brummel; A Chakilam; W.A Dorsch; B Fan; J.E Cochran; S Halas; E.M Harrington; J.K Hogan; D Howe; H Huang; D.H Jacobs; L.M Laitinen; S Liao; S Mahajan; Marone; G Martinez-Botella; P McCarthy; D Messersmith; Mark Namchuk; L Oh; M.S Penney; Albert Pierce; S.A Raybuck

While several therapeutic options exist, the need for more effective, safe, and convenient treatment for a variety of autoimmune diseases persists. Targeting the Janus tyrosine kinases (JAKs), which play essential roles in cell signaling responses and can contribute to aberrant immune function associated with disease, has emerged as a novel and attractive approach for the development of new autoimmune disease therapies. We screened our compound library against JAK3, a key signaling kinase in immune cells, and identified multiple scaffolds showing good inhibitory activity for this kinase. A particular scaffold of interest, the 1H-pyrrolo[2,3-b]pyridine series (7-azaindoles), was selected for further optimization in part on the basis of binding affinity (Ki) as well as on the basis of cellular potency. Optimization of this chemical series led to the identification of VX-509 (decernotinib), a novel, potent, and selective JAK3 inhibitor, which demonstrates good efficacy in vivo in the rat host versus graft model (HvG). On the basis of these findings, it appears that VX-509 offers potential for the treatment of a variety of autoimmune diseases.


Bioorganic & Medicinal Chemistry Letters | 2009

Structure-based design and parallel synthesis of N-benzyl isatin oximes as JNK3 MAP kinase inhibitors.

Jingrong Cao; Huai Gao; Guy W. Bemis; Francesco Salituro; Mark Ledeboer; Edmund Harrington; Susanne Wilke; Paul Taslimi; S. Pazhanisamy; Xiaoling Xie; Marc Jacobs; Jeremy Green

A series of N-benzylated isatin oximes were developed as inhibitors of the mitogen-activated kinase, JNK3. X-ray crystallographic structures aided in the design and synthesis of novel, selective compounds, that inhibit JNK3, but not p38 MAP kinase and provided key insights into understanding the behavior of gatekeeper residue methionine-146 in determining target selectivity for this series.


Bioorganic & Medicinal Chemistry Letters | 2009

2-Aminopyrazolo[1,5-a]pyrimidines as potent and selective inhibitors of JAK2.

Mark Ledeboer; Albert Pierce; John P. Duffy; Huai Gao; David Messersmith; Francesco Salituro; Suganthini Nanthakumar; Jon H. Come; Harmon Zuccola; Lora Swenson; Dina Shlyakter; Sudipta Mahajan; Thomas Hoock; Bin Fan; Wan-Jung Tsai; Elaine Kolaczkowski; Scott Carrier; James K. Hogan; Richard Zessis; S. Pazhanisamy; Youssef L. Bennani

Constitutive activation of the EPO/JAK2 signaling cascade has recently been implicated in a variety of myeloproliferative disorders including polycythemia vera, essential thrombocythemia and myelofibrosis. In an effort to uncover therapeutic potential of blocking the EPO/JAK2 signaling cascade, we sought to discover selective inhibitors that block the kinase activity of JAK2. Herein, we describe the discovery and structure based optimization of a novel series of 2-amino-pyrazolo[1,5-a]pyrimidines that exhibit potent inhibition of JAK2.


Bioorganic & Medicinal Chemistry Letters | 2015

Isosteric replacements of the carboxylic acid of drug candidate VX-787: Effect of charge on antiviral potency and kinase activity of azaindole-based influenza PB2 inhibitors.

Michael J. Boyd; Upul K. Bandarage; Hamilton Bennett; Randal R. Byrn; Ioana Davies; Wenxin Gu; Marc Jacobs; Mark Ledeboer; Brian Ledford; Joshua R. Leeman; Emanuele Perola; Tiansheng Wang; Youssef L. Bennani; Michael P. Clark; Paul S. Charifson

VX-787 is a first in class, orally bioavailable compound that offers unparalleled potential for the treatment of pandemic and seasonal influenza. As a part of our routine SAR exploration, carboxylic acid isosteres of VX-787 were prepared and tested against influenza A. It was found that the negative charge is important for maintaining potency and selectivity relative to kinase targets. Neutral carboxylic acid replacements generally resulted in compounds that were significantly less potent and less selective relative to the charged species.


Journal of Medicinal Chemistry | 2015

Discovery of Highly Isoform Selective Thiazolopiperidine Inhibitors of Phosphoinositide 3-Kinase γ

Philip N. Collier; David Messersmith; Arnaud Le Tiran; Upul K. Bandarage; Christina Boucher; Jon Come; Kevin M. Cottrell; Veronique Damagnez; John D. Doran; James P. Griffith; Suvarna Khare-Pandit; Elaine Krueger; Mark Ledeboer; Brian Ledford; Yusheng Liao; Sudipta Mahajan; Cameron Stuver Moody; Setu Roday; Tiansheng Wang; Jinwang Xu; Alex Aronov

A series of high affinity second-generation thiazolopiperidine inhibitors of PI3Kγ were designed based on some general observations around lipid kinase structure. Optimization of the alkylimidazole group led to inhibitors with higher levels of PI3Kγ selectivity. Additional insights into PI3K isoform selectivity related to sequence differences in a known distal hydrophobic pocket are also described.


ACS Medicinal Chemistry Letters | 2017

Novel 2-Substituted 7-Azaindole and 7-Azaindazole Analogues as Potential Antiviral Agents for the Treatment of Influenza

Upul K. Bandarage; Michael P. Clark; Emanuele Perola; Huai Gao; Marc Jacobs; Alice Tsai; Jeffery Gillespie; Joseph M. Kennedy; Francois Maltais; Mark Ledeboer; Ioana Davies; Wenxin Gu; Randal Byrn; Kwame Nti Addae; Hamilton Bennett; Joshua R. Leeman; Steven M. Jones; Colleen O’Brien; Christine Memmott; Youssef L. Bennani; Paul S. Charifson

JNJ-63623872 (2) is a first-in-class, orally bioavailable compound that offers significant potential for the treatment of pandemic and seasonal influenza. Early lead optimization efforts in our 7-azaindole series focused on 1,3-diaminocyclohexyl amide and urea substitutions on the pyrimidine-7-azaindole motif. In this work, we explored two strategies to eliminate observed aldehyde oxidase (AO)-mediated metabolism at the 2-position of these 7-azaindole analogues. Substitution at the 2-position of the azaindole ring generated somewhat less potent analogues, but reduced AO-mediated metabolism. Incorporation of a ring nitrogen generated 7-azaindazole analogues that were equipotent to the parent 2-H-7-azaindole, but surprisingly, did not appear to improve AO-mediated metabolism. Overall, we identified multiple 2-substituted 7-azaindole analogues with enhanced AO stability and we present data for one such compound (12) that demonstrate a favorable oral pharmacokinetic profile in rodents. These analogues have the potential to be further developed as anti-influenza agents for the treatment of influenza.

Collaboration


Dive into the Mark Ledeboer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Wang

Vertex Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huai Gao

Vertex Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge