Network


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

Hotspot


Dive into the research topics where Victor S. Gehling is active.

Publication


Featured researches published by Victor S. Gehling.


Chemistry & Biology | 2014

EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation.

William D. Bradley; Shilpi Arora; Jennifer Busby; Srividya Balasubramanian; Victor S. Gehling; Christopher G. Nasveschuk; Rishi G. Vaswani; Chih-Chi Yuan; Charlie Hatton; Feng Zhao; Kaylyn E. Williamson; Priyadarshini Iyer; Jacqui Mendez; Robert E. Campbell; Nico Cantone; Shivani Garapaty-Rao; James E. Audia; Andrew Simon Cook; Les A. Dakin; Brian K. Albrecht; Jean-Christophe Harmange; Danette L. Daniels; Richard T. Cummings; Barbara M. Bryant; Emmanuel Normant; Patrick Trojer

The histone lysine methyltransferase (MT) Enhancer of Zeste Homolog 2 (EZH2) is considered an oncogenic driver in a subset of germinal center B-cell-like diffuse large B cell lymphoma (GCB-DLBCL) and follicular lymphoma due to the presence of recurrent, monoallelic mutations in the EZH2 catalytic domain. These genomic data suggest that targeting the EZH2 MT activity is a valid therapeutic strategy for the treatment of lymphoma patients with EZH2 mutations. Here we report the identification of highly potent and selective EZH2 small molecule inhibitors, their validation by a cellular thermal shift assay, application across a large cell panel representing various non-Hodgkins lymphoma (NHL) subtypes, and their efficacy in EZH2mutant-containing GCB-DLBCL xenograft models. Surprisingly, our EZH2 inhibitors selectively affect the turnover of trimethylated, but not monomethylated histone H3 lysine 27 at pharmacologically relevant doses. Importantly, we find that these inhibitors are broadly efficacious also in NHL models with wild-type EZH2.


ACS Medicinal Chemistry Letters | 2013

Discovery, Design, and Optimization of Isoxazole Azepine BET Inhibitors.

Victor S. Gehling; Michael C. Hewitt; Rishi G. Vaswani; Yves Leblanc; Alexandre Côté; Christopher G. Nasveschuk; Alexander M. Taylor; Jean-Christophe Harmange; James E. Audia; Eneida Pardo; Shivangi Joshi; Peter Sandy; Jennifer A. Mertz; Robert J. Sims; Louise Bergeron; Barbara M. Bryant; Steve Bellon; Florence Poy; Hariharan Jayaram; Ravichandran Sankaranarayanan; Sreegouri Yellapantula; Nandana Bangalore Srinivasamurthy; Swarnakumari Birudukota; Brian K. Albrecht

The identification of a novel series of small molecule BET inhibitors is described. Using crystallographic binding modes of an amino-isoxazole fragment and known BET inhibitors, a structure-based drug design effort lead to a novel isoxazole azepine scaffold. This scaffold showed good potency in biochemical and cellular assays and oral activity in an in vivo model of BET inhibition.


Nature Chemical Biology | 2016

An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells

Maia Vinogradova; Victor S. Gehling; Amy Gustafson; Shilpi Arora; Charles Tindell; Catherine Wilson; Kaylyn E. Williamson; Gulfem D. Guler; Pranoti Gangurde; Wanda Manieri; Jennifer Busby; E. Megan Flynn; Fei Lan; Hyo-Jin Kim; Shobu Odate; Andrea G. Cochran; Yichin Liu; Matthew Wongchenko; Yibin Yang; Tommy K. Cheung; Tobias M. Maile; Ted Lau; Michael Costa; Ganapati V. Hegde; Erica Jackson; Robert M. Pitti; David Arnott; Christopher M. Bailey; Steve Bellon; Richard T. Cummings

The KDM5 family of histone demethylases catalyzes the demethylation of histone H3 on lysine 4 (H3K4) and is required for the survival of drug-tolerant persister cancer cells (DTPs). Here we report the discovery and characterization of the specific KDM5 inhibitor CPI-455. The crystal structure of KDM5A revealed the mechanism of inhibition of CPI-455 as well as the topological arrangements of protein domains that influence substrate binding. CPI-455 mediated KDM5 inhibition, elevated global levels of H3K4 trimethylation (H3K4me3) and decreased the number of DTPs in multiple cancer cell line models treated with standard chemotherapy or targeted agents. These findings show that pretreatment of cancer cells with a KDM5-specific inhibitor results in the ablation of a subpopulation of cancer cells that can serve as the founders for therapeutic relapse.


Journal of Medicinal Chemistry | 2016

Identification of a Benzoisoxazoloazepine Inhibitor (CPI-0610) of the Bromodomain and Extra-Terminal (BET) Family as a Candidate for Human Clinical Trials.

Brian K. Albrecht; Victor S. Gehling; Michael C. Hewitt; Rishi G. Vaswani; Alexandre Côté; Yves Leblanc; Christopher G. Nasveschuk; Steve Bellon; Louise Bergeron; Robert E. Campbell; Nico Cantone; Michael R. Cooper; Richard T. Cummings; Hariharan Jayaram; Shivangi Joshi; Jennifer A. Mertz; Adrianne Neiss; Emmanuel Normant; Michael O’Meara; Eneida Pardo; Florence Poy; Peter Sandy; Jeffrey G. Supko; Robert J. Sims; Jean-Christophe Harmange; Alexander M. Taylor; James E. Audia

In recent years, inhibition of the interaction between the bromodomain and extra-terminal domain (BET) family of chromatin adaptors and acetyl-lysine residues on chromatin has emerged as a promising approach to regulate the expression of important disease-relevant genes, including MYC, BCL-2, and NF-κB. Here we describe the identification and characterization of a potent and selective benzoisoxazoloazepine BET bromodomain inhibitor that attenuates BET-dependent gene expression in vivo, demonstrates antitumor efficacy in an MV-4-11 mouse xenograft model, and is currently undergoing human clinical trials for hematological malignancies (CPI-0610).


Journal of Medicinal Chemistry | 2016

Identification of (R)-N-((4-Methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide (CPI-1205), a Potent and Selective Inhibitor of Histone Methyltransferase EZH2, Suitable for Phase I Clinical Trials for B-Cell Lymphomas.

Rishi G. Vaswani; Victor S. Gehling; Les A. Dakin; Andrew Simon Cook; Christopher G. Nasveschuk; Martin Duplessis; Priyadarshini Iyer; Srividya Balasubramanian; Feng Zhao; Andrew C. Good; Robert E. Campbell; Christina R. Lee; Nico Cantone; Richard T. Cummings; Emmanuel Normant; Steven Bellon; Brian K. Albrecht; Jean-Christophe Harmange; Patrick Trojer; James E. Audia; Ying Zhang; Neil Justin; Shuyang Chen; Jon R. Wilson; Steven J. Gamblin

Polycomb repressive complex 2 (PRC2) has been shown to play a major role in transcriptional silencing in part by installing methylation marks on lysine 27 of histone 3. Dysregulation of PRC2 function correlates with certain malignancies and poor prognosis. EZH2 is the catalytic engine of the PRC2 complex and thus represents a key candidate oncology target for pharmacological intervention. Here we report the optimization of our indole-based EZH2 inhibitor series that led to the identification of CPI-1205, a highly potent (biochemical IC50 = 0.002 μM, cellular EC50 = 0.032 μM) and selective inhibitor of EZH2. This compound demonstrates robust antitumor effects in a Karpas-422 xenograft model when dosed at 160 mg/kg BID and is currently in Phase I clinical trials. Additionally, we disclose the co-crystal structure of our inhibitor series bound to the human PRC2 complex.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery, design, and synthesis of indole-based EZH2 inhibitors.

Victor S. Gehling; Rishi G. Vaswani; Christopher G. Nasveschuk; Martin Duplessis; Priyadarshini Iyer; Srividya Balasubramanian; Feng Zhao; Andrew C. Good; Robert E. Campbell; Christina R. Lee; Les A. Dakin; Andrew Simon Cook; Alexandre Gagnon; Jean-Christophe Harmange; James E. Audia; Richard T. Cummings; Emmanuel Normant; Patrick Trojer; Brian K. Albrecht

The discovery and optimization of a series of small molecule EZH2 inhibitors is described. Starting from dimethylpyridone HTS hit (2), a series of indole-based EZH2 inhibitors were identified. Biochemical potency and microsomal stability were optimized during these studies and afforded compound 22. This compound demonstrates nanomolar levels of biochemical potency (IC50=0.002 μM), cellular potency (EC50=0.080 μM), and afforded tumor regression when dosed (200 mpk SC BID) in an EZH2 dependent tumor xenograft model.


Organic Letters | 2014

A Practical Synthesis of Indoles via a Pd-Catalyzed C−N Ring Formation

Rishi G. Vaswani; Brian K. Albrecht; James E. Audia; Alexandre Côté; Les A. Dakin; Martin Duplessis; Victor S. Gehling; Jean-Christophe Harmange; Michael C. Hewitt; Yves Leblanc; Christopher G. Nasveschuk; Alexander M. Taylor

A method for the synthesis of N-functionalized C2-/C3-substituted indoles via Pd-catalyzed C-N bond coupling of halo-aryl enamines is described. The general strategy utilizes a variety of amines and β-keto esters which are elaborated into halo-aryl enamines as latent precursors to indoles. The preferred conditions comprising the RuPhos precatalyst and RuPhos in the presence of NaOMe in 1,4-dioxane tolerate a variety of substituents and are scalable for the construction of indoles in multigram quantities.


Bioorganic & Medicinal Chemistry Letters | 2016

Identification of potent, selective KDM5 inhibitors

Victor S. Gehling; Steven Bellon; Jean-Christophe Harmange; Yves Leblanc; Florence Poy; Shobu Odate; Shane Buker; Fei Lan; Shilpi Arora; Kaylyn E. Williamson; Peter Sandy; Richard T. Cummings; Christopher M. Bailey; Louise Bergeron; Weifeng Mao; Amy Gustafson; Yichin Liu; Erica VanderPorten; James E. Audia; Patrick Trojer; Brian K. Albrecht

This communication describes the identification and optimization of a series of pan-KDM5 inhibitors derived from compound 1, a hit initially identified against KDM4C. Compound 1 was optimized to afford compound 20, a 10nM inhibitor of KDM5A. Compound 20 is highly selective for the KDM5 enzymes versus other histone lysine demethylases and demonstrates activity in a cellular assay measuring the increase in global histone 3 lysine 4 tri-methylation (H3K4me3). In addition compound 20 has good ADME properties, excellent mouse PK, and is a suitable starting point for further optimization.


Bioorganic & Medicinal Chemistry Letters | 2016

Lead optimization of a pyrazolo[1,5-a]pyrimidin-7(4H)-one scaffold to identify potent, selective and orally bioavailable KDM5 inhibitors suitable for in vivo biological studies.

Jun Liang; Birong Zhang; Sharada Labadie; Daniel F. Ortwine; Maia Vinogradova; James R. Kiefer; Victor S. Gehling; Jean-Christophe Harmange; Richard D. Cummings; Tommy Lai; Jiangpeng Liao; Xiaoping Zheng; Yichin Liu; Amy Gustafson; Erica Van der Porten; Weifeng Mao; Bianca M. Liederer; Gauri Deshmukh; Marie Classon; Patrick Trojer; Peter S. Dragovich; Lesley J. Murray

Starting with a lead [1,5-a]pyrimidin-7(4H)-one-containing molecule (1), we generated potent, selective and orally bioavailable KDM5 inhibitors. Using structure- and property-based approaches, we designed 48 with improved cell potency (PC9 H3K4Me3 EC50=0.34μM). Furthermore, 48 maintained suitable physiochemical properties and displayed an excellent pharmacokinetic (PK) profile in mice. When dosed orally in mice at 50mg/kg twice a day (BID), 48 showed an unbound maximal plasma concentration (Cmax) >15-fold over its cell EC50, thereby providing a robust chemical probe for studying KDM5 biological functions in vivo.


Bioorganic & Medicinal Chemistry Letters | 2015

Development of methyl isoxazoleazepines as inhibitors of BET

Michael C. Hewitt; Yves Leblanc; Victor S. Gehling; Rishi G. Vaswani; Alexandre Côté; Christopher G. Nasveschuk; Alexander M. Taylor; Jean-Christophe Harmange; James E. Audia; Eneida Pardo; Rich Cummings; Shivangi Joshi; Peter Sandy; Jennifer A. Mertz; Robert J. Sims; Louise Bergeron; Barbara M. Bryant; Steve Bellon; Florence Poy; Hariharan Jayaram; Yong Tang; Brian K. Albrecht

In this report we detail the evolution of our previously reported thiophene isoxazole BET inhibitor chemotype exemplified by CPI-3 to a novel bromodomain selective chemotype (the methyl isoxazoleazepine chemotype) exemplified by carboxamide 23. The methyl isoxazoleazepine chemotype provides potent inhibition of the bromodomains of the BET family, excellent in vivo PK across species, low unbound clearance, and target engagement in a MYC PK-PD model.

Collaboration


Dive into the Victor S. Gehling's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael C. Hewitt

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge