Network


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

Hotspot


Dive into the research topics where Damien Y. Duveau is active.

Publication


Featured researches published by Damien Y. Duveau.


Proceedings of the National Academy of Sciences of the United States of America | 2014

High-throughput combinatorial screening identifies drugs that cooperate with ibrutinib to kill activated B-cell-like diffuse large B-cell lymphoma cells.

Lesley A. Mathews Griner; Rajarshi Guha; Paul Shinn; Ryan M. Young; Jonathan M. Keller; Dongbo Liu; Ian S. Goldlust; Adam Yasgar; Crystal McKnight; Matthew B. Boxer; Damien Y. Duveau; Jian-kang Jiang; Sam Michael; Tim Mierzwa; Wenwei Huang; Martin J. Walsh; Bryan T. Mott; Paresma R. Patel; William Leister; David J. Maloney; Christopher A. LeClair; Ganesha Rai; Ajit Jadhav; Brian D. Peyser; Christopher P. Austin; Scott E. Martin; Anton Simeonov; Marc Ferrer; Louis M. Staudt; Craig J. Thomas

Significance The treatment of cancer is highly reliant on drug combinations. Next-generation, targeted therapeutics are demonstrating interesting single-agent activities in clinical trials; however, the discovery of companion drugs through iterative clinical trial-and-error is not a tenable mechanism to prioritize clinically important combinations for these agents. Herein we describe the results of a large, high-throughput combination screen of the Bruton’s tyrosine kinase inhibitor ibrutinib versus a library of nearly 500 approved and investigational drugs. Multiple ibrutinib combinations were discovered through this study that can be prioritized for clinical examination. The clinical development of drug combinations is typically achieved through trial-and-error or via insight gained through a detailed molecular understanding of dysregulated signaling pathways in a specific cancer type. Unbiased small-molecule combination (matrix) screening represents a high-throughput means to explore hundreds and even thousands of drug–drug pairs for potential investigation and translation. Here, we describe a high-throughput screening platform capable of testing compounds in pairwise matrix blocks for the rapid and systematic identification of synergistic, additive, and antagonistic drug combinations. We use this platform to define potential therapeutic combinations for the activated B-cell–like subtype (ABC) of diffuse large B-cell lymphoma (DLBCL). We identify drugs with synergy, additivity, and antagonism with the Bruton’s tyrosine kinase inhibitor ibrutinib, which targets the chronic active B-cell receptor signaling that characterizes ABC DLBCL. Ibrutinib interacted favorably with a wide range of compounds, including inhibitors of the PI3K-AKT-mammalian target of rapamycin signaling cascade, other B-cell receptor pathway inhibitors, Bcl-2 family inhibitors, and several components of chemotherapy that is the standard of care for DLBCL.


ACS Chemical Biology | 2015

A small molecule that inhibits OGT activity in cells.

Rodrigo F. Ortiz-Meoz; Jiaoyang Jiang; Michael Lazarus; Marina Orman; John Janetzko; Chenguang Fan; Damien Y. Duveau; Zhi Wei Tan; Craig J. Thomas; Suzanne Walker

O-GlcNAc transferase (OGT) is an essential mammalian enzyme that regulates numerous cellular processes through the attachment of O-linked N-acetylglucosamine (O-GlcNAc) residues to nuclear and cytoplasmic proteins. Its targets include kinases, phosphatases, transcription factors, histones, and many other intracellular proteins. The biology of O-GlcNAc modification is still not well understood, and cell-permeable inhibitors of OGT are needed both as research tools and for validating OGT as a therapeutic target. Here, we report a small molecule OGT inhibitor, OSMI-1, developed from a high-throughput screening hit. It is cell-permeable and inhibits protein O-GlcNAcylation in several mammalian cell lines without qualitatively altering cell surface N- or O-linked glycans. The development of this molecule validates high-throughput screening approaches for the discovery of glycosyltransferase inhibitors, and further optimization of this scaffold may lead to yet more potent OGT inhibitors useful for studying OGT in animal models.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Selective targeting of JAK/STAT signaling is potentiated by Bcl-xL blockade in IL-2–dependent adult T-cell leukemia

Meili Zhang; Lesley A. Mathews Griner; Wei Ju; Damien Y. Duveau; Rajarshi Guha; Michael N. Petrus; Bernard Wen; Michiyuki Maeda; Paul Shinn; Marc Ferrer; Kevin D. Conlon; Richard Bamford; John J. O’Shea; Craig J. Thomas; Thomas A. Waldmann

Significance Adult T-cell leukemia (ATL) is caused by the human T-cell lymphotropic virus-1 (HTLV-1). Presently there is no curative therapy for ATL. In the leukemic cells, the HTLV-1–encoded protein Tax (transactivator from the X-gene region) activates disordered interleukin expression, which triggers JAK/STAT signaling. The HTLV-1–encoded protein Tax also induces the expression of the antiapoptotic protein Bcl-xL (B-cell lymphoma-extra large). In the present study, the combination of the JAK inhibitor ruxolitinib and the Bcl-2/Bcl-xL inhibitor navitoclax provided additive/synergistic antitumor efficacy in IL-2–dependent ATL cell lines and in a mouse model of human ATL as well as with ex vivo peripheral blood mononuclear cells (PBMCs) from ATL patients compared with either drug alone, supporting a therapeutic trial of this combination in patients with ATL. Adult T-cell leukemia (ATL) develops in individuals infected with human T-cell lymphotropic virus-1 (HTLV-1). Presently there is no curative therapy for ATL. HTLV-1–encoded protein Tax (transactivator from the X-gene region) up-regulates Bcl-xL (B-cell lymphoma-extra large) expression and activates interleukin-2 (IL-2), IL-9, and IL-15 autocrine/paracrine systems, resulting in amplified JAK/STAT signaling. Inhibition of JAK signaling reduces cytokine-dependent ex vivo proliferation of peripheral blood mononuclear cells (PBMCs) from ATL patients in smoldering/chronic stages. Currently, two JAK inhibitors are approved for human use. In this study, we examined activity of multiple JAK inhibitors in ATL cell lines. The selective JAK inhibitor ruxolitinib was examined in a high-throughput matrix screen combined with >450 potential therapeutic agents, and Bcl-2/Bcl-xL inhibitor navitoclax was identified as a strong candidate for multicomponent therapy. The combination was noted to strongly activate BAX (Bcl-2-associated X protein), effect mitochondrial depolarization, and increase caspase 3/7 activities that lead to cleavage of PARP (poly ADP ribose polymerase) and Mcl-1 (myeloid cell leukemia 1). Ruxolitinib and navitoclax independently demonstrated modest antitumor efficacy, whereas the combination dramatically lowered tumor burden and prolonged survival in an ATL murine model. This combination strongly blocked ex vivo proliferation of five ATL patients’ PBMCs. These studies provide support for a therapeutic trial in patients with smoldering/chronic ATL using a drug combination that inhibits JAK signaling and antiapoptotic protein Bcl-xL.


Nature | 2017

Targeting neuronal activity-regulated neuroligin-3 dependency in high-grade glioma

Humsa Venkatesh; Lydia T. Tam; Pamelyn Woo; James Lennon; Surya Nagaraja; Shawn M. Gillespie; Jing Ni; Damien Y. Duveau; Patrick J. Morris; Jean Zhao; Craig J. Thomas; Michelle Monje

High-grade gliomas (HGG) are a devastating group of cancers, and represent the leading cause of brain tumour-related death in both children and adults. Therapies aimed at mechanisms intrinsic to glioma cells have translated to only limited success; effective therapeutic strategies will need also to target elements of the tumour microenvironment that promote glioma progression. Neuronal activity promotes the growth of a range of molecularly and clinically distinct HGG types, including adult and paediatric glioblastoma (GBM), anaplastic oligodendroglioma, and diffuse intrinsic pontine glioma (DIPG). An important mechanism that mediates this neural regulation of brain cancer is activity-dependent cleavage and secretion of the synaptic adhesion molecule neuroligin-3 (NLGN3), which promotes glioma proliferation through the PI3K–mTOR pathway. However, the necessity of NLGN3 for glioma growth, the proteolytic mechanism of NLGN3 secretion, and the further molecular consequences of NLGN3 secretion in glioma cells remain unknown. Here we show that HGG growth depends on microenvironmental NLGN3, identify signalling cascades downstream of NLGN3 binding in glioma, and determine a therapeutically targetable mechanism of secretion. Patient-derived orthotopic xenografts of paediatric GBM, DIPG and adult GBM fail to grow in Nlgn3 knockout mice. NLGN3 stimulates several oncogenic pathways, such as early focal adhesion kinase activation upstream of PI3K–mTOR, and induces transcriptional changes that include upregulation of several synapse-related genes in glioma cells. NLGN3 is cleaved from both neurons and oligodendrocyte precursor cells via the ADAM10 sheddase. ADAM10 inhibitors prevent the release of NLGN3 into the tumour microenvironment and robustly block HGG xenograft growth. This work defines a promising strategy for targeting NLGN3 secretion, which could prove transformative for HGG therapy.


Bioorganic & Medicinal Chemistry Letters | 2013

Synthesis and biological evaluation of analogues of the kinase inhibitor nilotinib as Abl and Kit inhibitors.

Damien Y. Duveau; Xin Hu; Martin J. Walsh; Suneet Shukla; Amanda P. Skoumbourdis; Matthew B. Boxer; Suresh V. Ambudkar; Min Shen; Craig J. Thomas

The importance of the trifluoromethyl group in the polypharmacological profile of nilotinib was investigated. Molecular editing of nilotinib led to the design, synthesis and biological evaluation of analogues where the trifluoromethyl group was replaced by a proton, fluorine and a methyl group. While these analogues were less active than nilotinib toward Abl, their activity toward Kit was comparable, with the monofluorinated analogue being the most active. Docking of nilotinib and of analogues 2a-c to the binding pocket of Abl and of Kit showed that the lack of shape complementarity in Kit is compensated by the stabilizing effect from its juxtamembrane region.


Leukemia | 2014

Elucidation of the structural basis of interaction of the BCR-ABL kinase inhibitor, nilotinib (Tasigna) with the human ABC drug transporter P-glycoprotein

Suneet Shukla; Eduardo E. Chufan; Satyakam Singh; Amanda P. Skoumbourdis; Khyati Kapoor; Matthew B. Boxer; Damien Y. Duveau; Craig J. Thomas; Tanaji T. Talele; Suresh V. Ambudkar

Elucidation of the structural basis of interaction of the BCR-ABL kinase inhibitor, nilotinib (Tasigna) with the human ABC drug transporter P-glycoprotein


Oncotarget | 2016

Inhibition of O-GlcNAc transferase activity reprograms prostate cancer cell metabolism

Harri Itkonen; Saurabh Sayajirao Gorad; Damien Y. Duveau; Sara E. S. Martin; Anna Barkovskaya; Tone F. Bathen; Siver A. Moestue; Ian G. Mills

Metabolic networks are highly connected and complex, but a single enzyme, O-GlcNAc transferase (OGT) can sense the availability of metabolites and also modify target proteins. We show that inhibition of OGT activity inhibits the proliferation of prostate cancer cells, leads to sustained loss of c-MYC and suppresses the expression of CDK1, elevated expression of which predicts prostate cancer recurrence (p=0.00179). Metabolic profiling revealed decreased glucose consumption and lactate production after OGT inhibition. This decreased glycolytic activity specifically sensitized prostate cancer cells, but not cells representing normal prostate epithelium, to inhibitors of oxidative phosphorylation (rotenone and metformin). Intra-cellular alanine was depleted upon OGT inhibitor treatment. OGT inhibitor increased the expression and activity of alanine aminotransferase (GPT2), an enzyme that can be targeted with a clinically approved drug, cycloserine. Simultaneous inhibition of OGT and GPT2 inhibited cell viability and growth rate, and additionally activated a cell death response. These combinatorial effects were predominantly seen in prostate cancer cells, but not in a cell-line derived from normal prostate epithelium. Combinatorial treatments were confirmed with two inhibitors against both OGT and GPT2. Taken together, here we report the reprogramming of energy metabolism upon inhibition of OGT activity, and identify synergistically lethal combinations that are prostate cancer cell specific.


Bioorganic & Medicinal Chemistry Letters | 2014

Structure-activity relationship studies and biological characterization of human NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase inhibitors.

Damien Y. Duveau; Adam Yasgar; Yuhong Wang; Xin Hu; Jennifer Kouznetsova; Kyle R. Brimacombe; Ajit Jadhav; Anton Simeonov; Craig J. Thomas; David J. Maloney

The structure-activity relationship (SAR) study of two chemotypes identified as inhibitors of the human NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase (HPGD, 15-PGDH) was conducted. Top compounds from both series displayed potent inhibition (IC50 <50 nM), demonstrate excellent selectivity towards HPGD and potently induce PGE2 production in A549 lung cancer and LNCaP prostate cancer cells.


Clinical Cancer Research | 2017

Matrix screen identifies synergistic combination of PARP inhibitors and nicotinamide phosphoribosyltransferase (NAMPT) inhibitors in Ewing sarcoma

Christine Heske; Mindy I. Davis; Joshua T. Baumgart; Kelli Wilson; Michael Gormally; Lu Chen; Xiaohu Zhang; Michele Ceribelli; Damien Y. Duveau; Rajarshi Guha; Marc Ferrer; Fernanda I. Arnaldez; Jiuping Ji; Huong Lan Tran; Yiping Zhang; Arnulfo Mendoza; Lee J. Helman; Craig J. Thomas

Purpose: Although many cancers are showing remarkable responses to targeted therapies, pediatric sarcomas, including Ewing sarcoma, remain recalcitrant. To broaden the therapeutic landscape, we explored the in vitro response of Ewing sarcoma cell lines against a large collection of investigational and approved drugs to identify candidate combinations. Experimental Design: Drugs displaying activity as single agents were evaluated in combinatorial (matrix) format to identify highly active, synergistic drug combinations, and combinations were subsequently validated in multiple cell lines using various agents from each class. Comprehensive metabolomic and proteomic profiling was performed to better understand the mechanism underlying the synergy. Xenograft experiments were performed to determine efficacy and in vivo mechanism. Results: Several promising candidates emerged, including the combination of small-molecule PARP and nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, a rational combination as NAMPTis block the rate-limiting enzyme in the production of nicotinamide adenine dinucleotide (NAD+), a necessary substrate of PARP. Mechanistic drivers of the synergistic cell killing phenotype of these combined drugs included depletion of NMN and NAD+, diminished PAR activity, increased DNA damage, and apoptosis. Combination PARPis and NAMPTis in vivo resulted in tumor regression, delayed disease progression, and increased survival. Conclusions: These studies highlight the potential of these drugs as a possible therapeutic option in treating patients with Ewing sarcoma. Clin Cancer Res; 23(23); 7301–11. ©2017 AACR.


Journal of the American Chemical Society | 2018

Structure-Based Evolution of Low Nanomolar O-GlcNAc Transferase Inhibitors.

Sara E. S. Martin; Zhi-Wei Tan; Harri M. Itkonen; Damien Y. Duveau; Joao A. Paulo; John Janetzko; Paul L. Boutz; Lisa Törk; Frederick A. Moss; Craig J. Thomas; Steven P. Gygi; Michael B. Lazarus; Suzanne Walker

Reversible glycosylation of nuclear and cytoplasmic proteins is an important regulatory mechanism across metazoans. One enzyme, O-linked N-acetylglucosamine transferase (OGT), is responsible for all nucleocytoplasmic glycosylation and there is a well-known need for potent, cell-permeable inhibitors to interrogate OGT function. Here we report the structure-based evolution of OGT inhibitors culminating in compounds with low nanomolar inhibitory potency and on-target cellular activity. In addition to disclosing useful OGT inhibitors, the structures we report provide insight into how to inhibit glycosyltransferases, a family of enzymes that has been notoriously refractory to inhibitor development.

Collaboration


Dive into the Damien Y. Duveau's collaboration.

Top Co-Authors

Avatar

Craig J. Thomas

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marc Ferrer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Adam Yasgar

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ajit Jadhav

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Anton Simeonov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David J. Maloney

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xiaohu Zhang

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Xin Hu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jennifer Kouznetsova

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kyle R. Brimacombe

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge