Network


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

Hotspot


Dive into the research topics where Matthew B. Boxer is active.

Publication


Featured researches published by Matthew B. Boxer.


Science | 2011

Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses

Dimitrios Anastasiou; George Poulogiannis; John M. Asara; Matthew B. Boxer; Jian-kang Jiang; Min Shen; Gary Bellinger; Atsuo T. Sasaki; Jason W. Locasale; Douglas S. Auld; Craig J. Thomas; Matthew G. Vander Heiden; Lewis C. Cantley

The glycolytic metabolism of cancers differs from normal tissues, allowing tumor cells to survive under oxidative stress. Control of intracellular reactive oxygen species (ROS) concentrations is critical for cancer cell survival. We show that, in human lung cancer cells, acute increases in intracellular concentrations of ROS caused inhibition of the glycolytic enzyme pyruvate kinase M2 (PKM2) through oxidation of Cys358. This inhibition of PKM2 is required to divert glucose flux into the pentose phosphate pathway and thereby generate sufficient reducing potential for detoxification of ROS. Lung cancer cells in which endogenous PKM2 was replaced with the Cys358 to Ser358 oxidation-resistant mutant exhibited increased sensitivity to oxidative stress and impaired tumor formation in a xenograft model. Besides promoting metabolic changes required for proliferation, the regulatory properties of PKM2 may confer an additional advantage to cancer cells by allowing them to withstand oxidative stress.


Nature Chemical Biology | 2012

Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis

Dimitrios Anastasiou; Yimin Yu; William J. Israelsen; Jian Kang Jiang; Matthew B. Boxer; Bum Soo Hong; Wolfram Tempel; Svetoslav Dimov; Min Shen; Abhishek K. Jha; Hua Yang; Katherine R. Mattaini; Christian M. Metallo; Brian Prescott Fiske; Kevin D. Courtney; Scott Malstrom; Tahsin M. Khan; Charles Kung; Amanda P. Skoumbourdis; Henrike Veith; Noel Southall; Martin J. Walsh; Kyle R. Brimacombe; William Leister; Sophia Y. Lunt; Zachary R. Johnson; Katharine E. Yen; Kaiko Kunii; Shawn M. Davidson; Heather R. Christofk

Cancer cells engage in a metabolic program to enhance biosynthesis and support cell proliferation. The regulatory properties of pyruvate kinase M2 (PKM2) influence altered glucose metabolism in cancer. PKM2 interaction with phosphotyrosine-containing proteins inhibits enzyme activity and increases availability of glycolytic metabolites to support cell proliferation. This suggests that high pyruvate kinase activity may suppress tumor growth. We show that expression of PKM1, the pyruvate kinase isoform with high constitutive activity, or exposure to published small molecule PKM2 activators inhibit growth of xenograft tumors. Structural studies reveal that small molecule activators bind PKM2 at the subunit interaction interface, a site distinct from that of the endogenous activator fructose-1,6-bisphosphate (FBP). However, unlike FBP, binding of activators to PKM2 promotes a constitutively active enzyme state that is resistant to inhibition by tyrosine-phosphorylated proteins. These data support the notion that small molecule activation of PKM2 can interfere with anabolic metabolism.


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.


Nature | 2015

Drug-based modulation of endogenous stem cells promotes functional remyelination in vivo

Fadi J. Najm; Mayur Madhavan; Anita Zaremba; Elizabeth Shick; Robert T. Karl; Daniel C. Factor; Tyler E. Miller; Zachary S. Nevin; Christopher Kantor; Alex Sargent; Kevin L. Quick; Daniela Schlatzer; Hong Tang; Ruben Papoian; Kyle R. Brimacombe; Min Shen; Matthew B. Boxer; Ajit Jadhav; Andrew P. Robinson; Joseph R. Podojil; Stephen D. Miller; Robert H. Miller; Paul J. Tesar

Multiple sclerosis involves an aberrant autoimmune response and progressive failure of remyelination in the central nervous system. Prevention of neural degeneration and subsequent disability requires remyelination through the generation of new oligodendrocytes, but current treatments exclusively target the immune system. Oligodendrocyte progenitor cells are stem cells in the central nervous system and the principal source of myelinating oligodendrocytes. These cells are abundant in demyelinated regions of patients with multiple sclerosis, yet fail to differentiate, thereby representing a cellular target for pharmacological intervention. To discover therapeutic compounds for enhancing myelination from endogenous oligodendrocyte progenitor cells, we screened a library of bioactive small molecules on mouse pluripotent epiblast stem-cell-derived oligodendrocyte progenitor cells. Here we show seven drugs function at nanomolar doses selectively to enhance the generation of mature oligodendrocytes from progenitor cells in vitro. Two drugs, miconazole and clobetasol, are effective in promoting precocious myelination in organotypic cerebellar slice cultures, and in vivo in early postnatal mouse pups. Systemic delivery of each of the two drugs significantly increases the number of new oligodendrocytes and enhances remyelination in a lysolecithin-induced mouse model of focal demyelination. Administering each of the two drugs at the peak of disease in an experimental autoimmune encephalomyelitis mouse model of chronic progressive multiple sclerosis results in striking reversal of disease severity. Immune response assays show that miconazole functions directly as a remyelinating drug with no effect on the immune system, whereas clobetasol is a potent immunosuppressant as well as a remyelinating agent. Mechanistic studies show that miconazole and clobetasol function in oligodendrocyte progenitor cells through mitogen-activated protein kinase and glucocorticoid receptor signalling, respectively. Furthermore, both drugs enhance the generation of human oligodendrocytes from human oligodendrocyte progenitor cells in vitro. Collectively, our results provide a rationale for testing miconazole and clobetasol, or structurally modified derivatives, to enhance remyelination in patients.


Bioorganic & Medicinal Chemistry Letters | 2010

Evaluation of thieno[3,2-b]pyrrole[3,2-d]pyridazinones as activators of the tumor cell specific M2 isoform of pyruvate kinase

Jian-kang Jiang; Matthew B. Boxer; Matthew G. Vander Heiden; Min Shen; Amanda P. Skoumbourdis; Noel Southall; Henrike Veith; William Leister; Christopher P. Austin; Hee-Won Park; James Inglese; Lewis C. Cantley; Douglas S. Auld; Craig J. Thomas

Cancer cells have distinct metabolic needs that are different from normal cells and can be exploited for development of anti-cancer therapeutics. Activation of the tumor specific M2 form of pyruvate kinase (PKM2) is a potential strategy for returning cancer cells to a metabolic state characteristic of normal cells. Here, we describe activators of PKM2 based upon a substituted thieno[3,2-b]pyrrole[3,2-d]pyridazinone scaffold. The synthesis of these agents, structure-activity relationships, analysis of activity at related targets (PKM1, PKR and PKL) and examination of aqueous solubility are investigated. These agents represent the second reported chemotype for activation of PKM2.


Nature Communications | 2015

Quantitative high throughput screening using a primary human three-dimensional organotypic culture predicts in vivo efficacy

Hilary A. Kenny; Madhu Lal-Nag; Erin A. White; Min Shen; Chun Yi Chiang; Anirban K. Mitra; Yilin Zhang; Marion Curtis; Elizabeth M. Schryver; Sam Bettis; Ajit Jadhav; Matthew B. Boxer; Zhuyin Li; Marc Ferrer; Ernst Lengyel

The tumour microenvironment contributes to cancer metastasis and drug resistance. However, most high throughput screening (HTS) assays for drug discovery use cancer cells grown in monolayers. Here we show that a multilayered culture containing primary human fibroblasts, mesothelial cells and extracellular matrix can be adapted into a reliable 384- and 1,536-multi-well HTS assay that reproduces the human ovarian cancer (OvCa) metastatic microenvironment. We validate the identified inhibitors in secondary in vitro and in vivo biological assays using three OvCa cell lines: HeyA8, SKOV3ip1 and Tyk-nu. The active compounds directly inhibit at least two of the three OvCa functions: adhesion, invasion and growth. In vivo, these compounds prevent OvCa adhesion, invasion and metastasis, and improve survival in mouse models. Collectively, these data indicate that a complex three-dimensional culture of the tumour microenvironment can be adapted for quantitative HTS and may improve the disease relevance of assays used for drug screening.


Nature Chemical Biology | 2016

A PHGDH inhibitor reveals coordination of serine synthesis and one-carbon unit fate

Michael E. Pacold; Kyle R. Brimacombe; Sze Ham Chan; Jason M. Rohde; Caroline A. Lewis; Lotteke J.Y.M. Swier; Richard Possemato; Walter W. Chen; Lucas B. Sullivan; Brian Prescott Fiske; Sung Won Cho; Elizaveta Freinkman; Kivanc Birsoy; Monther Abu-Remaileh; Yoav D. Shaul; Chieh Min Liu; Minerva Zhou; Min Jung Koh; Haeyoon Chung; Shawn M. Davidson; Alba Luengo; Amy Wang; Xin Xu; Adam Yasgar; Li Liu; Ganesha Rai; Kenneth D. Westover; Matthew G. Vander Heiden; Min Shen; Nathanael S. Gray

Serine is a both a proteinogenic amino acid and the source of one-carbon units essential for de novo purine and deoxythymidine synthesis. In the canonical glucose-derived serine synthesis pathway, Homo sapiens phosphoglycerate dehydrogenase (PHGDH) catalyzes the first, rate-limiting step. Genetic loss of PHGDH is toxic towards PHGDH-overexpressing breast cancer cell lines even in the presence of exogenous serine. Here, we use a quantitative high-throughput screen to identify small molecule PHGDH inhibitors. These compounds reduce the production of glucose-derived serine in cells and suppress the growth of PHGDH-dependent cancer cells in culture and in orthotopic xenograft tumors. Surprisingly, PHGDH inhibition reduced the incorporation into nucleotides of one-carbon units from glucose-derived and exogenous serine. We conclude that glycolytic serine synthesis coordinates the use of one-carbon units from endogenous and exogenous serine in nucleotide synthesis, and suggest that one-carbon unit wasting may contribute to the efficacy of PHGDH inhibitors in vitro and in vivo.


Bioorganic & Medicinal Chemistry Letters | 2011

2-oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamides as activators of the tumor cell specific M2 isoform of pyruvate kinase

Martin J. Walsh; Kyle R. Brimacombe; Henrike Veith; James M. Bougie; Thomas Oran Daniel; William Leister; Lewis C. Cantley; William J. Israelsen; Matthew G. Vander Heiden; Min Shen; Douglas S. Auld; Craig J. Thomas; Matthew B. Boxer

Compared to normal differentiated cells, cancer cells have altered metabolic regulation to support biosynthesis and the expression of the M2 isozyme of pyruvate kinase (PKM2) plays an important role in this anabolic metabolism. While the M1 isoform is a highly active enzyme, the alternatively spliced M2 variant is considerably less active and expressed in tumors. While the exact mechanism by which decreased pyruvate kinase activity contributes to anabolic metabolism remains unclear, it is hypothesized that activation of PKM2 to levels seen with PKM1 may promote a metabolic program that is not conducive to cell proliferation. Here we report the third chemotype in a series of PKM2 activators based on the 2-oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamide scaffold. The synthesis, structure activity relationships, selectivity and notable physiochemical properties are described.


Journal of Biological Chemistry | 2014

Biochemical, Cellular and Biophysical Characterization of a Potent Inhibitor of Mutant Isocitrate Dehydrogenase IDH1

Mindy I. Davis; Stefan Gross; Min Shen; Kimberly Straley; Rajan Pragani; Wendy A. Lea; Janeta Popovici-Muller; Byron DeLaBarre; Erin Artin; Natasha Thorne; Douglas S. Auld; Zhuyin Li; Lenny Dang; Matthew B. Boxer; Anton Simeonov

Background: IDH1 R132H, implicated in glioblastoma and AML, produces the oncometabolite 2-HG. Results: A detailed binding mechanism of a small molecule inhibitor (ML309) is proposed. Conclusion: ML309 competes with α-KG but is uncompetitive with NADPH and rapidly and reversibly affects cellular 2-HG levels. Significance: Understanding IDH1 R132H inhibition sets the stage for targeting IDH1 R132H for the treatment of cancer. Two mutant forms (R132H and R132C) of isocitrate dehydrogenase 1 (IDH1) have been associated with a number of cancers including glioblastoma and acute myeloid leukemia. These mutations confer a neomorphic activity of 2-hydroxyglutarate (2-HG) production, and 2-HG has previously been implicated as an oncometabolite. Inhibitors of mutant IDH1 can potentially be used to treat these diseases. In this study, we investigated the mechanism of action of a newly discovered inhibitor, ML309, using biochemical, cellular, and biophysical approaches. Substrate binding and product inhibition studies helped to further elucidate the IDH1 R132H catalytic cycle. This rapidly equilibrating inhibitor is active in both biochemical and cellular assays. The (+) isomer is active (IC50 = 68 nm), whereas the (−) isomer is over 400-fold less active (IC50 = 29 μm) for IDH1 R132H inhibition. IDH1 R132C was similarly inhibited by (+)-ML309. WT IDH1 was largely unaffected by (+)-ML309 (IC50 >36 μm). Kinetic analyses combined with microscale thermophoresis and surface plasmon resonance indicate that this reversible inhibitor binds to IDH1 R132H competitively with respect to α-ketoglutarate and uncompetitively with respect to NADPH. A reaction scheme for IDH1 R132H inhibition by ML309 is proposed in which ML309 binds to IDH1 R132H after formation of the IDH1 R132H NADPH complex. ML309 was also able to inhibit 2-HG production in a glioblastoma cell line (IC50 = 250 nm) and had minimal cytotoxicity. In the presence of racemic ML309, 2-HG levels drop rapidly. This drop was sustained until 48 h, at which point the compound was washed out and 2-HG levels recovered.


Molecular Cell | 2015

PEPCK Coordinates the Regulation of Central Carbon Metabolism to Promote Cancer Cell Growth

Emily Montal; Ruby Dewi; Kavita Bhalla; Lihui Ou; Bor Jang Hwang; Ashley E. Ropell; Chris Gordon; Wan Ju Liu; Ralph J. DeBerardinis; Jessica Sudderth; William Twaddel; Laszlo G. Boros; Kenneth R. Shroyer; Sekhar Duraisamy; Ronny Drapkin; R. Scott Powers; Jason M. Rohde; Matthew B. Boxer; Kwok-Kin Wong; Geoffrey D. Girnun

Phosphoenolpyruvate carboxykinase (PEPCK) is well known for its role in gluconeogenesis. However, PEPCK is also a key regulator of TCA cycle flux. The TCA cycle integrates glucose, amino acid, and lipid metabolism depending on cellular needs. In addition, biosynthetic pathways crucial to tumor growth require the TCA cycle for the processing of glucose and glutamine derived carbons. We show here an unexpected role for PEPCK in promoting cancer cell proliferation in vitro and in vivo by increasing glucose and glutamine utilization toward anabolic metabolism. Unexpectedly, PEPCK also increased the synthesis of ribose from non-carbohydrate sources, such as glutamine, a phenomenon not previously described. Finally, we show that the effects of PEPCK on glucose metabolism and cell proliferation are in part mediated via activation of mTORC1. Taken together, these data demonstrate a role for PEPCK that links metabolic flux and anabolic pathways to cancer cell proliferation.

Collaboration


Dive into the Matthew B. Boxer's collaboration.

Top Co-Authors

Avatar

Min Shen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Douglas S. Auld

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Craig J. Thomas

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kyle R. Brimacombe

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

Martin J. Walsh

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Matthew G. Vander Heiden

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Henrike Veith

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian-kang Jiang

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge