Network


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

Hotspot


Dive into the research topics where Robert A. Copeland is active.

Publication


Featured researches published by Robert A. Copeland.


Journal of Biological Chemistry | 1998

Identification of a Novel Inhibitor of Mitogen-activated Protein Kinase Kinase

Margaret F. Favata; Kurumi Y. Horiuchi; Elizabeth J. Manos; Andrea J. Daulerio; Deborah A. Stradley; Wendi S. Feeser; Drew E. Van Dyk; William John Pitts; Richard A. Earl; Frank W. Hobbs; Robert A. Copeland; Ronald L. Magolda; Peggy Scherle; James M. Trzaskos

The compound U0126 (1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio]butadiene) was identified as an inhibitor of AP-1 transactivation in a cell-based reporter assay. U0126 was also shown to inhibit endogenous promoters containing AP-1 response elements but did not affect genes lacking an AP-1 response element in their promoters. These effects of U0126 result from direct inhibition of the mitogen-activated protein kinase kinase family members, MEK-1 and MEK-2. Inhibition is selective for MEK-1 and -2, as U0126 shows little, if any, effect on the kinase activities of protein kinase C, Abl, Raf, MEKK, ERK, JNK, MKK-3, MKK-4/SEK, MKK-6, Cdk2, or Cdk4. Comparative kinetic analysis of U0126 and the MEK inhibitor PD098059 (Dudley, D. T., Pang, L., Decker, S. J., Bridges, A. J., and Saltiel, A. R. (1995) Proc. Natl. Acad. Sci U. S. A. 92, 7686–7689) demonstrates that U0126 and PD098059 are noncompetitive inhibitors with respect to both MEK substrates, ATP and ERK. We further demonstrate that the two compounds bind to ΔN3-S218E/S222D MEK in a mutually exclusive fashion, suggesting that they may share a common or overlapping binding site(s). Quantitative evaluation of the steady state kinetics of MEK inhibition by these compounds reveals that U0126 has approximately 100-fold higher affinity for ΔN3-S218E/S222D MEK than does PD098059. We further tested the effects of these compounds on the activity of wild type MEK isolated after activation from stimulated cells. Surprisingly, we observe a significant diminution in affinity of both compounds for wild type MEK as compared with the ΔN3-S218E/S222D mutant enzyme. These results suggest that the affinity of both compounds is mediated by subtle conformational differences between the two activated MEK forms. The MEK affinity of U0126, its selectivity for MEK over other kinases, and its cellular efficacy suggest that this compound will serve as a powerful tool for in vitro and cellular investigations of mitogen-activated protein kinase-mediated signal transduction.


Nature Chemical Biology | 2012

A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells

Sarah K. Knutson; Tim J. Wigle; Natalie Warholic; Christopher John Sneeringer; Christina J. Allain; Christine R. Klaus; Joelle D Sacks; Alejandra Raimondi; Christina R. Majer; Jeffrey Song; Margaret Porter Scott; Lei Jin; Jesse J. Smith; Edward J. Olhava; Richard Chesworth; Mikel P. Moyer; Victoria M. Richon; Robert A. Copeland; Heike Keilhack; Roy M. Pollock; Kevin Wayne Kuntz

EZH2 catalyzes trimethylation of histone H3 lysine 27 (H3K27). Point mutations of EZH2 at Tyr641 and Ala677 occur in subpopulations of non-Hodgkins lymphoma, where they drive H3K27 hypertrimethylation. Here we report the discovery of EPZ005687, a potent inhibitor of EZH2 (K(i) of 24 nM). EPZ005687 has greater than 500-fold selectivity against 15 other protein methyltransferases and has 50-fold selectivity against the closely related enzyme EZH1. The compound reduces H3K27 methylation in various lymphoma cells; this translates into apoptotic cell killing in heterozygous Tyr641 or Ala677 mutant cells, with minimal effects on the proliferation of wild-type cells. These data suggest that genetic alteration of EZH2 (for example, mutations at Tyr641 or Ala677) results in a critical dependency on enzymatic activity for proliferation (that is, the equivalent of oncogene addiction), thus portending the clinical use of EZH2 inhibitors for cancers in which EZH2 is genetically altered.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Coordinated activities of wild-type plus mutant EZH2 drive tumor-associated hypertrimethylation of lysine 27 on histone H3 (H3K27) in human B-cell lymphomas

Christopher John Sneeringer; Margaret Porter Scott; Kevin Wayne Kuntz; Sarah K. Knutson; Roy M. Pollock; Victoria M. Richon; Robert A. Copeland

EZH2, the catalytic subunit of the PRC2 complex, catalyzes the mono- through trimethylation of lysine 27 on histone H3 (H3K27). Histone H3K27 trimethylation is a mechanism for suppressing transcription of specific genes that are proximal to the site of histone modification. Point mutations of the EZH2 gene (Tyr641) have been reported to be linked to subsets of human B-cell lymphoma. The mutant allele is always found associated with a wild-type allele (heterozygous) in disease cells, and the mutations were reported to ablate the enzymatic activity of the PRC2 complex for methylating an unmodified peptide substrate. Here we demonstrate that the WT enzyme displays greatest catalytic efficiency (kcat/K) for the zero to monomethylation reaction of H3K27 and diminished efficiency for subsequent (mono- to di- and di- to trimethylation) reactions. In stark contrast, the disease-associated Y641 mutations display very limited ability to perform the first methylation reaction, but have enhanced catalytic efficiency for the subsequent reactions, relative to the WT enzyme. These results imply that the malignant phenotype of disease requires the combined activities of a H3K27 monomethylating enzyme (PRC2 containing WT EZH2 or EZH1) together with the mutant PRC2s for augmented conversion of H3K27 to the trimethylated form. To our knowledge, this is the first example of a human disease that is dependent on the coordinated activities of normal and disease-associated mutant enzymatic function.


Biochemistry | 2008

Residence Time of Receptor-Ligand Complexes and Its Effect on Biological Function

Peter J. Tummino; Robert A. Copeland

The formation and duration of binary receptor-ligand complexes are fundamental to many physiologic processes. Most often, the effectiveness of interaction between a receptor and its ligand is quantified in terms of closed system, equilibrium affinity measurements, such as IC50 and Kd. In the context of in vivo biology, however, the extent and duration of responses to receptor-ligand interactions depend greatly on the time period over which the ligand is in residence on its receptor. Here we define receptor-ligand complex residence time in quantitative terms and describe its significance to biological function. Examples of the importance of residence time are presented for natural ligands of different receptor types. The impact of residence time on the optimization of potential ligands as drugs for human medicine is also described.


Bioorganic & Medicinal Chemistry Letters | 1998

MEK inhibitors: the chemistry and biological activity of U0126, its analogs, and cyclization products.

John V. Duncia; Joseph B. Santella; C. Anne Higley; William John Pitts; John Wityak; William E. Frietze; F.Wayne Rankin; Jung-Hui Sun; Richard A. Earl; A.Christine Tabaka; Christopher A. Teleha; Karl F. Blom; Margaret F. Favata; Elizabeth J. Manos; Andrea J. Daulerio; Deborah A. Stradley; Kurumi Y. Horiuchi; Robert A. Copeland; Peggy Scherle; James M. Trzaskos; Ronald L. Magolda; George L. Trainor; Ruth R. Wexler; Frank W. Hobbs; Richard E. Olson

In search of antiinflammatory drugs with a new mechanism of action, U0126 was found to functionally antagonize AP-1 transcriptional activity via noncompetitive inhibition of the dual specificity kinase MEK with an IC50 of 0.07 microM for MEK 1 and 0.06 microM for MEK 2. U0126 can undergo isomerization and cyclization reactions to form a variety of products, both chemically and in vivo, all of which exhibit less affinity for MEK and lower inhibition of AP-1 activity than parent, U0126.


Journal of Biological Chemistry | 2000

Presenilin-1 and 2 are molecular targets for gamma secretase inhibitors

Dietmar A. Seiffert; Jodi D. Bradley; Cynthia M. Rominger; David H. Rominger; Fude Yang; Jere E. Meredith; Qian Wang; Arthur H. Roach; Lorin A. Thompson; Susan M. Spitz; Jeffrey N. Higaki; Shimoga R. Prakash; Andrew P. Combs; Robert A. Copeland; Stephen P. Arneric; Paul R. Hartig; David W. Robertson; Barbara Cordell; Andrew M. Stern; Richard E. Olson; Robert Zaczek

Presenilins are integral membrane protein involved in the production of amyloid β-protein. Mutations of the presenilin-1 and -2 gene are associated with familial Alzheimers disease and are thought to alter γ-secretase cleavage of the β-amyloid precursor protein, leading to increased production of longer and more amyloidogenic forms of Aβ, the 4-kDa β-peptide. Here, we show that radiolabeled γ-secretase inhibitors bind to mammalian cell membranes, and a benzophenone analog specifically photocross-links three major membrane polypeptides. A positive correlation is observed among these compounds for inhibition of cellular Aβ formation, inhibition of membrane binding and cross-linking. Immunological techniques establish N- and C-terminal fragments of presenilin-1 as specifically cross-linked polypeptides. Furthermore, binding of γ-secretase inhibitors to embryonic membranes derived from presenilin-1 knockout embryos is reduced in a gene dose-dependent manner. In addition, C-terminal fragments of presenilin-2 are specifically cross-linked. Taken together, these results indicate that potent and selective γ-secretase inhibitors block Aβ formation by binding to presenilin-1 and -2.


Blood | 2013

Potent inhibition of DOT1L as treatment of MLL-fusion leukemia

Scott R. Daigle; Edward J. Olhava; Carly A. Therkelsen; Aravind Basavapathruni; Lei Jin; P. Ann Boriack-Sjodin; Christina J. Allain; Christine R. Klaus; Alejandra Raimondi; Margaret Porter Scott; Nigel J. Waters; Richard Chesworth; Mikel P. Moyer; Robert A. Copeland; Victoria M. Richon; Roy M. Pollock

Rearrangements of the MLL gene define a genetically distinct subset of acute leukemias with poor prognosis. Current treatment options are of limited effectiveness; thus, there is a pressing need for new therapies for this disease. Genetic and small molecule inhibitor studies have demonstrated that the histone methyltransferase DOT1L is required for the development and maintenance of MLL-rearranged leukemia in model systems. Here we describe the characterization of EPZ-5676, a potent and selective aminonucleoside inhibitor of DOT1L histone methyltransferase activity. The compound has an inhibition constant value of 80 pM, and demonstrates 37 000-fold selectivity over all other methyltransferases tested. In cellular studies, EPZ-5676 inhibited H3K79 methylation and MLL-fusion target gene expression and demonstrated potent cell killing that was selective for acute leukemia lines bearing MLL translocations. Continuous IV infusion of EPZ-5676 in a rat xenograft model of MLL-rearranged leukemia caused complete tumor regressions that were sustained well beyond the compound infusion period with no significant weight loss or signs of toxicity. EPZ-5676 is therefore a potential treatment of MLL-rearranged leukemia and is under clinical investigation.


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

Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2

Sarah K. Knutson; Natalie Warholic; Tim J. Wigle; Christine R. Klaus; Christina J. Allain; Alejandra Raimondi; Margaret Porter Scott; Richard Chesworth; Mikel P. Moyer; Robert A. Copeland; Victoria M. Richon; Roy M. Pollock; Kevin Wayne Kuntz; Heike Keilhack

Inactivation of the switch/sucrose nonfermentable complex component SMARCB1 is extremely prevalent in pediatric malignant rhabdoid tumors (MRTs) or atypical teratoid rhabdoid tumors. This alteration is hypothesized to confer oncogenic dependency on EZH2 in these cancers. We report the discovery of a potent, selective, and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity, (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[1,1′-biphenyl]-3-carboxamide). The compound induces apoptosis and differentiation specifically in SMARCB1-deleted MRT cells. Treatment of xenograft-bearing mice with (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4′-(morpholinomethyl)-[1,1′-biphenyl]-3-carboxamide) leads to dose-dependent regression of MRTs with correlative diminution of intratumoral trimethylation levels of lysine 27 on histone H3, and prevention of tumor regrowth after dosing cessation. These data demonstrate the dependency of SMARCB1 mutant MRTs on EZH2 enzymatic activity and portend the utility of EZH2-targeted drugs for the treatment of these genetically defined cancers.


Nature Chemical Biology | 2015

The promise and peril of chemical probes

C.H. Arrowsmith; James E. Audia; Christopher M. Austin; Jonathan B. Baell; Jonathan Bennett; Julian Blagg; C. Bountra; Paul E. Brennan; Peter J. Brown; Mark Edward Bunnage; Carolyn Buser-Doepner; Robert M. Campbell; Adrian Carter; Philip Cohen; Robert A. Copeland; Ben Cravatt; Jayme L. Dahlin; Dashyant Dhanak; A. Edwards; Mathias Frederiksen; Stephen V. Frye; Nathanael S. Gray; Charles E. Grimshaw; David Hepworth; Trevor Howe; Kilian Huber; Jian Jin; Stefan Knapp; Joanne Kotz; Ryan G. Kruger

Chemical probes are powerful reagents with increasing impacts on biomedical research. However, probes of poor quality or that are used incorrectly generate misleading results. To help address these shortcomings, we will create a community-driven wiki resource to improve quality and convey current best practice.


Molecular Cancer Therapeutics | 2014

Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma

Sarah K. Knutson; Satoshi Kawano; Yukinori Minoshima; Natalie Warholic; Kuan-Chun Huang; Yonghong Xiao; Tadashi Kadowaki; Mai Uesugi; Galina Kuznetsov; Namita Kumar; Tim J. Wigle; Christine R. Klaus; Christina J. Allain; Alejandra Raimondi; Nigel J. Waters; Jesse J. Smith; Margaret Porter-Scott; Richard Chesworth; Mikel P. Moyer; Robert A. Copeland; Victoria M. Richon; Toshimitsu Uenaka; Roy M. Pollock; Kevin Wayne Kuntz; Akira Yokoi; Heike Keilhack

Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers. Mol Cancer Ther; 13(4); 842–54. ©2014 AACR.

Collaboration


Dive into the Robert A. Copeland's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tim J. Wigle

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge