Network


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

Hotspot


Dive into the research topics where Sharad K. Verma is active.

Publication


Featured researches published by Sharad K. Verma.


Nature | 2012

EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations

Michael T. McCabe; Heidi M. Ott; Gopinath Ganji; Susan Korenchuk; Christine Thompson; Glenn S. Van Aller; Yan Liu; Alan P. Graves; Anthony Della Pietra; Elsie Diaz; Louis V. LaFrance; Mark Mellinger; Celine Duquenne; Xinrong Tian; Ryan G. Kruger; Charles F. McHugh; Martin Brandt; William Henry Miller; Dashyant Dhanak; Sharad K. Verma; Peter J. Tummino; Caretha L. Creasy

In eukaryotes, post-translational modification of histones is critical for regulation of chromatin structure and gene expression. EZH2 is the catalytic subunit of the polycomb repressive complex 2 (PRC2) and is involved in repressing gene expression through methylation of histone H3 on lysine 27 (H3K27). EZH2 overexpression is implicated in tumorigenesis and correlates with poor prognosis in several tumour types. Additionally, somatic heterozygous mutations of Y641 and A677 residues within the catalytic SET domain of EZH2 occur in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma. The Y641 residue is the most frequently mutated residue, with up to 22% of germinal centre B-cell DLBCL and follicular lymphoma harbouring mutations at this site. These lymphomas have increased H3K27 tri-methylation (H3K27me3) owing to altered substrate preferences of the mutant enzymes. However, it is unknown whether specific, direct inhibition of EZH2 methyltransferase activity will be effective in treating EZH2 mutant lymphomas. Here we demonstrate that GSK126, a potent, highly selective, S-adenosyl-methionine-competitive, small-molecule inhibitor of EZH2 methyltransferase activity, decreases global H3K27me3 levels and reactivates silenced PRC2 target genes. GSK126 effectively inhibits the proliferation of EZH2 mutant DLBCL cell lines and markedly inhibits the growth of EZH2 mutant DLBCL xenografts in mice. Together, these data demonstrate that pharmacological inhibition of EZH2 activity may provide a promising treatment for EZH2 mutant lymphoma.


Cancer Cell | 2013

EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation

Wendy Béguelin; Relja Popovic; Matt Teater; Yanwen Jiang; Karen L. Bunting; Monica Rosen; Hao Shen; Shao Ning Yang; Ling Wang; Teresa Ezponda; Eva Martinez-Garcia; Haikuo Zhang; Sharad K. Verma; Michael T. McCabe; Heidi M. Ott; Glenn S. Van Aller; Ryan G. Kruger; Yan Liu; Charles F. McHugh; David W. Scott; Young Rock Chung; Neil L. Kelleher; Rita Shaknovich; Caretha L. Creasy; Randy D. Gascoyne; Kwok-Kin Wong; Leandro Cerchietti; Ross L. Levine; Omar Abdel-Wahab; Jonathan D. Licht

The EZH2 histone methyltransferase is highly expressed in germinal center (GC) B cells and targeted by somatic mutations in B cell lymphomas. Here, we find that EZH2 deletion or pharmacologic inhibition suppresses GC formation and functions. EZH2 represses proliferation checkpoint genes and helps establish bivalent chromatin domains at key regulatory loci to transiently suppress GC B cell differentiation. Somatic mutations reinforce these physiological effects through enhanced silencing of EZH2 targets. Conditional expression of mutant EZH2 in mice induces GC hyperplasia and accelerated lymphomagenesis in cooperation with BCL2. GC B cell (GCB)-type diffuse large B cell lymphomas (DLBCLs) are mostly addicted to EZH2 but not the more differentiated activated B cell (ABC)-type DLBCLs, thus clarifying the therapeutic scope of EZH2 targeting.


Nature | 2014

Epigenomic alterations define lethal CIMP-positive ependymomas of infancy.

Stephen C. Mack; Hendrik Witt; Rosario M. Piro; Lei Gu; Scott Zuyderduyn; A. M. Stütz; Xiaosong Wang; Marco Gallo; Livia Garzia; Kory Zayne; Xiaoyang Zhang; Vijay Ramaswamy; Natalie Jäger; David T. W. Jones; Martin Sill; Trevor J. Pugh; M. Ryzhova; Khalida Wani; David Shih; Renee Head; Marc Remke; S. D. Bailey; Thomas Zichner; Claudia C. Faria; Mark Barszczyk; Sebastian Stark; Huriye Seker-Cin; Sonja Hutter; Pascal Johann; Sebastian Bender

Ependymomas are common childhood brain tumours that occur throughout the nervous system, but are most common in the paediatric hindbrain. Current standard therapy comprises surgery and radiation, but not cytotoxic chemotherapy as it does not further increase survival. Whole-genome and whole-exome sequencing of 47 hindbrain ependymomas reveals an extremely low mutation rate, and zero significant recurrent somatic single nucleotide variants. Although devoid of recurrent single nucleotide variants and focal copy number aberrations, poor-prognosis hindbrain ependymomas exhibit a CpG island methylator phenotype. Transcriptional silencing driven by CpG methylation converges exclusively on targets of the Polycomb repressive complex 2 which represses expression of differentiation genes through trimethylation of H3K27. CpG island methylator phenotype-positive hindbrain ependymomas are responsive to clinical drugs that target either DNA or H3K27 methylation both in vitro and in vivo. We conclude that epigenetic modifiers are the first rational therapeutic candidates for this deadly malignancy, which is epigenetically deregulated but genetically bland.


ACS Medicinal Chemistry Letters | 2012

Identification of Potent, Selective, Cell-Active Inhibitors of the Histone Lysine Methyltransferase EZH2.

Sharad K. Verma; Xinrong Tian; Louis Vincent Lafrance; Celine Duquenne; Dominic Suarez; Kenneth A. Newlander; Stuart P. Romeril; Joelle L. Burgess; Seth W. Grant; James Brackley; Alan P. Graves; Daryl Scherzer; Art Shu; Christine Thompson; Heidi M. Ott; Glenn S. Van Aller; Carl A. Machutta; Elsie Diaz; Yong Jiang; Neil W. Johnson; Steven David Knight; Ryan G. Kruger; Michael T. McCabe; Dashyant Dhanak; Peter J. Tummino; Caretha L. Creasy; William H. Miller

The histone H3-lysine 27 (H3K27) methyltransferase EZH2 plays a critical role in regulating gene expression, and its aberrant activity is linked to the onset and progression of cancer. As part of a drug discovery program targeting EZH2, we have identified highly potent, selective, SAM-competitive, and cell-active EZH2 inhibitors, including GSK926 (3) and GSK343 (6). These compounds are small molecule chemical tools that would be useful to further explore the biology of EZH2.


Journal of Biomolecular Screening | 2012

Development and Validation of Reagents and Assays for EZH2 Peptide and Nucleosome High-Throughput Screens

Elsie Diaz; Carl A. Machutta; Stephanie Chen; Yong Jiang; Christopher J. Nixon; Glenn A. Hofmann; Danielle Key; Sharon Sweitzer; Mehul Patel; Zining Wu; Caretha L. Creasy; Ryan G. Kruger; Louis V. LaFrance; Sharad K. Verma; Melissa B. Pappalardi; BaoChau Le; Glenn S. Van Aller; Michael T. McCabe; Peter J. Tummino; Andrew J. Pope; Sara H. Thrall; Benjamin Schwartz; Martin Brandt

Histone methyltransferases (HMT) catalyze the methylation of histone tail lysines, resulting in changes in gene transcription. Misregulation of these enzymes has been associated with various forms of cancer, making this target class a potential new area for the development of novel chemotherapeutics. EZH2 is the catalytic component of the polycomb group repressive complex (PRC2), which selectively methylates histone H3 lysine 27 (H3K27). EZH2 is overexpressed in prostate, breast, bladder, brain, and other tumor types and is recognized as a molecular marker for cancer progression and aggressiveness. Several new reagents and assays were developed to aid in the identification of EZH2 inhibitors, and these were used to execute two high-throughput screening campaigns. Activity assays using either an H3K27 peptide or nucleosomes as substrates for methylation are described. The strategy to screen EZH2 with either a surrogate peptide or a natural substrate led to the identification of the same tractable series. Compounds from this series are reversible, are [3H]-S-adenosyl-L-methionine competitive, and display biochemical inhibition of H3K27 methylation.


ACS Chemical Biology | 2014

Long residence time inhibition of EZH2 in activated polycomb repressive complex 2.

Glenn S. Van Aller; Melissa B. Pappalardi; Heidi M. Ott; Elsie Diaz; Martin Brandt; Benjamin J. Schwartz; William H. Miller; Dashyant Dhanak; Michael T. McCabe; Sharad K. Verma; Caretha L. Creasy; Peter J. Tummino; Ryan G. Kruger

EZH2/PRC2 catalyzes transcriptionally repressive methylation at lysine 27 of histone H3 and has been associated with numerous cancer types. Point mutations in EZH2 at Tyr641 and Ala677 identified in non-Hodgkin lymphomas alter substrate specificity and result in increased trimethylation at histone H3K27. Interestingly, EZH2/PRC2 is activated by binding H3K27me3 marks on histones, and this activation is proposed as a mechanism for self-propagation of gene silencing. Recent work has identified GSK126 as a potent, selective, SAM-competitive inhibitor of EZH2 capable of globally decreasing H3K27 trimethylation in cells. Here we show that activation of PRC2 by an H3 peptide trimethylated at K27 is primarily an effect on the rate-limiting step (kcat) with no effect on substrate binding (Km). Additionally, GSK126 is shown to have a significantly longer residence time of inhibition on the activated form of EZH2/PRC2 as compared to unactivated EZH2/PRC2. Overall inhibition constant (Ki*) values for GSK126 were determined to be as low as 93 pM and appear to be driven by slow dissociation of inhibitor from the activated enzyme. The data suggest that activation of EZH2 allows the enzyme to adopt a conformation that possesses greater affinity for GSK126. The long residence time of GSK126 may be beneficial in vivo and may result in durable target inhibition after drug systemic clearance.


Molecular Cancer Therapeutics | 2014

A687V EZH2 Is a Driver of Histone H3 Lysine 27 (H3K27) Hypertrimethylation

Heidi M. Ott; Alan P. Graves; Melissa B. Pappalardi; Michael Huddleston; Wendy S. Halsey; Ashley M. Hughes; Arthur Groy; Edward Dul; Yong Jiang; Yuchen Bai; Roland S. Annan; Sharad K. Verma; Steven D. Knight; Ryan G. Kruger; Dashyant Dhanak; Benjamin Schwartz; Peter J. Tummino; Caretha L. Creasy; Michael T. McCabe

The EZH2 methyltransferase silences gene expression through methylation of histone H3 on lysine 27 (H3K27). Recently, EZH2 mutations have been reported at Y641, A677, and A687 in non-Hodgkin lymphoma. Although the Y641F/N/S/H/C and A677G mutations exhibit clearly increased activity with substrates dimethylated at lysine 27 (H3K27me2), the A687V mutant has been shown to prefer a monomethylated lysine 27 (H3K27me1) with little gain of activity toward H3K27me2. Herein, we demonstrate that despite this unique substrate preference, A687V EZH2 still drives increased H3K27me3 when transiently expressed in cells. However, unlike the previously described mutants that dramatically deplete global H3K27me2 levels, A687V EZH2 retains normal levels of H3K27me2. Sequencing of B-cell–derived cancer cell lines identified an acute lymphoblastic leukemia cell line harboring this mutation. Similar to exogenous expression of A687V EZH2, this cell line exhibited elevated H3K27me3 while possessing H3K27me2 levels higher than Y641- or A677-mutant lines. Treatment of A687V EZH2-mutant cells with GSK126, a selective EZH2 inhibitor, was associated with a global decrease in H3K27me3, robust gene activation, caspase activation, and decreased proliferation. Structural modeling of the A687V EZH2 active site suggests that the increased catalytic activity with H3K27me1 may be due to a weakened interaction with an active site water molecule that must be displaced for dimethylation to occur. These findings suggest that A687V EZH2 likely increases global H3K27me3 indirectly through increased catalytic activity with H3K27me1 and cells harboring this mutation are highly dependent on EZH2 activity for their survival. Mol Cancer Ther; 13(12); 3062–73. ©2014 AACR.


Annual Reports in Medicinal Chemistry | 2007

Chapter 23 Small Molecule Inhibitors of AKT/PKB Kinase as a Strategy for Treating Cancer

Dirk A. Heerding; Igor G. Safonov; Sharad K. Verma

Publisher Summary This chapter discusses small molecule inhibitors of AKT/PKB kinase as a strategy for treating cancer. In studies the 2-pyrimidyl-5-amidothiophene has been identified as an ATP-competitive AKT inhibitor. Also the AKT activity of the quinoline sulfonamide analog derived from the PKA inhibitor H-89 has been disclosed in the literature. Perifosine is a recently found inhibitor which is in clinical trial. Perifosine is a synthetic, orally available alkylphospholipid, derived from alkylphosphocholine, which targets the PI3/AKT survival pathway. Although the molecular mechanism underlying the antineoplastic activity of perifosine is not fully elucidated, studies suggest that perifosine interferes with turnover and synthesis of natural phospholipids. This disrupts membrane signaling at several sites resulting in the inhibition of the PI3K/AKT survival pathway. Recent preclinical evaluation in cultured human Jurkat T-leukemia cells has shown that adding low concentrations of 27 (5 mM) after treatment with the commonly used chemotherapy drug etoposide, induced cell death in a synergistic fashion. The observed increase in cell death is attributed to an inactivation of the AKT survival pathway, as treated cells showed a complete dephosphorylation of AKT. Compound perifosine also inhibited baseline phosphorylation of AKT in multiple myeloma cells in a time- and dose-dependent fashion.


Bioorganic & Medicinal Chemistry Letters | 2010

2,3,5-Trisubstituted pyridines as selective AKT inhibitors. Part II: Improved drug-like properties and kinase selectivity from azaindazoles

Hong Lin; Dennis S. Yamashita; Jin Zeng; Ren Xie; Sharad K. Verma; Juan I. Luengo; Nelson Rhodes; Shu-Yun Zhang; Kimberly A. Robell; Anthony E. Choudhry; Zhihong Lai; Rakesh Kumar; Elisabeth A. Minthorn; Kristin K. Brown; Dirk A. Heerding

A novel series of AKT inhibitors containing 2,3,5-trisubstituted pyridines with novel azaindazoles as hinge binding elements are described. Among these, the 4,7-diazaindazole compound 2c has improved drug-like properties and kinase selectivity than those of indazole 1, and displays greater than 80% inhibition of GSK3beta phosphorylation in a BT474 tumor xenograft model in mice.


Future Medicinal Chemistry | 2013

Recent progress in the discovery of small-molecule inhibitors of the HMT EZH2 for the treatment of cancer

Sharad K. Verma; Steven D. Knight

The histone lysine methyltransferase EZH2 is the catalytic component of the multi-protein PRC2 complex and methylates lysine 27 on histone H3. EZH2 overexpression is implicated in tumorigenesis and correlates with poor prognosis in several tumor types. Inhibition of aberrant EZH2 activity might attenuate tumorigenesis resulting from misregulated gene transcription derived from aberrant EZH2 activity. In the last year, the first reports of small molecules demonstrating potent and selective inhibition of EZH2 have been published by multiple groups. Herein, we review recent progress reported in the discovery of small molecule inhibitors of EZH2.

Collaboration


Dive into the Sharad K. Verma'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge