Network


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

Hotspot


Dive into the research topics where Benito Munoz is active.

Publication


Featured researches published by Benito Munoz.


Cell | 2013

An Interactive Resource to Identify Cancer Genetic and Lineage Dependencies Targeted by Small Molecules

Amrita Basu; Nicole E. Bodycombe; Jaime H. Cheah; Edmund V. Price; Ke Liu; Giannina Ines Schaefer; Richard Yon Ebright; Michelle L. Stewart; Daisuke Ito; Stephanie Wang; Abigail L. Bracha; Ted Liefeld; Mathias J. Wawer; Joshua C. Gilbert; Andrew J. Wilson; Nicolas Stransky; Gregory V. Kryukov; Vlado Dančík; Jordi Barretina; Levi A. Garraway; C. Suk-Yee Hon; Benito Munoz; Joshua Bittker; Brent R. Stockwell; Dineo Khabele; Paul A. Clemons; Alykhan F. Shamji; Stuart L. Schreiber

The high rate of clinical response to protein-kinase-targeting drugs matched to cancer patients with specific genomic alterations has prompted efforts to use cancer cell line (CCL) profiling to identify additional biomarkers of small-molecule sensitivities. We have quantitatively measured the sensitivity of 242 genomically characterized CCLs to an Informer Set of 354 small molecules that target many nodes in cell circuitry, uncovering protein dependencies that: (1) associate with specific cancer-genomic alterations and (2) can be targeted by small molecules. We have created the Cancer Therapeutics Response Portal (http://www.broadinstitute.org/ctrp) to enable users to correlate genetic features to sensitivity in individual lineages and control for confounding factors of CCL profiling. We report a candidate dependency, associating activating mutations in the oncogene β-catenin with sensitivity to the Bcl-2 family antagonist, navitoclax. The resource can be used to develop novel therapeutic hypotheses and to accelerate discovery of drugs matched to patients by their cancer genotype and lineage.


Cancer Discovery | 2015

Harnessing Connectivity in a Large-Scale Small-Molecule Sensitivity Dataset

Brinton Seashore-Ludlow; Matthew G. Rees; Jaime H. Cheah; Murat Cokol; Edmund V. Price; Matthew E. Coletti; Victor Victor Jones; Nicole E. Bodycombe; Christian K. Soule; Joshua Gould; Benjamin Alexander; Ava Li; Philip Montgomery; Mathias J. Wawer; Nurdan Kuru; Joanne Kotz; C. Suk-Yee Hon; Benito Munoz; Ted Liefeld; Vlado Dančík; Joshua Bittker; Michelle Palmer; James E. Bradner; Alykhan F. Shamji; Paul A. Clemons; Stuart L. Schreiber

UNLABELLED Identifying genetic alterations that prime a cancer cell to respond to a particular therapeutic agent can facilitate the development of precision cancer medicines. Cancer cell-line (CCL) profiling of small-molecule sensitivity has emerged as an unbiased method to assess the relationships between genetic or cellular features of CCLs and small-molecule response. Here, we developed annotated cluster multidimensional enrichment analysis to explore the associations between groups of small molecules and groups of CCLs in a new, quantitative sensitivity dataset. This analysis reveals insights into small-molecule mechanisms of action, and genomic features that associate with CCL response to small-molecule treatment. We are able to recapitulate known relationships between FDA-approved therapies and cancer dependencies and to uncover new relationships, including for KRAS-mutant cancers and neuroblastoma. To enable the cancer community to explore these data, and to generate novel hypotheses, we created an updated version of the Cancer Therapeutic Response Portal (CTRP v2). SIGNIFICANCE We present the largest CCL sensitivity dataset yet available, and an analysis method integrating information from multiple CCLs and multiple small molecules to identify CCL response predictors robustly. We updated the CTRP to enable the cancer research community to leverage these data and analyses.


Journal of Biological Chemistry | 2010

Novel Inhibitors of Plasmodium falciparum Dihydroorotate Dehydrogenase with Anti-malarial Activity in the Mouse Model

Michael Booker; Cecilia M. Bastos; Martin Kramer; Robert Barker; Renato Skerlj; Amar Bir Singh Sidhu; Xiaoyi Deng; Cassandra Celatka; Joseph F. Cortese; Jose E. Guerrero Bravo; Keila N. Crespo Llado; Adelfa E. Serrano; Iñigo Angulo-Barturen; María Belén Jiménez-Díaz; Sara Viera; Helen Garuti; Sergio Wittlin; Petros Papastogiannidis; Jing-wen Lin; Chris J. Janse; Shahid M. Khan; Manoj T. Duraisingh; Bradley I. Coleman; Elizabeth J. Goldsmith; Margaret A. Phillips; Benito Munoz; Dyann F. Wirth; Jeffrey D. Klinger; Roger Wiegand; Edmund Sybertz

Plasmodium falciparum, the causative agent of the most deadly form of human malaria, is unable to salvage pyrimidines and must rely on de novo biosynthesis for survival. Dihydroorotate dehydrogenase (DHODH) catalyzes the rate-limiting step in the pyrimidine biosynthetic pathway and represents a potential target for anti-malarial therapy. A high throughput screen and subsequent medicinal chemistry program identified a series of N-alkyl-5-(1H-benzimidazol-1-yl)thiophene-2-carboxamides with low nanomolar in vitro potency against DHODH from P. falciparum, P. vivax, and P. berghei. The compounds were selective for the parasite enzymes over human DHODH, and x-ray structural data on the analog Genz-667348, demonstrated that species selectivity could be attributed to amino acid differences in the inhibitor-binding site. Compounds from this series demonstrated in vitro potency against the 3D7 and Dd2 strains of P. falciparum, good tolerability and oral exposure in the mouse, and ED50 values in the 4-day murine P. berghei efficacy model of 13–21 mg/kg/day with oral twice-daily dosing. In particular, treatment with Genz-667348 at 100 mg/kg/day resulted in sterile cure. Two recent analogs of Genz-667348 are currently undergoing pilot toxicity testing to determine suitability as clinical development candidates.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of a small-molecule inhibitor and cellular probe of Keap1-Nrf2 protein-protein interaction.

Longqin Hu; Sadagopan Magesh; Lin Chen; Lili Wang; Tim Lewis; Yu Chen; Carol Khodier; Daigo Inoyama; Lesa J. Beamer; Thomas J. Emge; Jian Shen; John E. Kerrigan; Ah-Ng Tony Kong; Sivaraman Dandapani; Michelle Palmer; Stuart L. Schreiber; Benito Munoz

A high-throughput screen (HTS) of the MLPCN library using a homogenous fluorescence polarization assay identified a small molecule as a first-in-class direct inhibitor of Keap1-Nrf2 protein-protein interaction. The HTS hit has three chiral centers; a combination of flash and chiral chromatographic separation demonstrated that Keap1-binding activity resides predominantly in one stereoisomer (SRS)-5 designated as ML334 (LH601A), which is at least 100× more potent than the other stereoisomers. The stereochemistry of the four cis isomers was assigned using X-ray crystallography and confirmed using stereospecific synthesis. (SRS)-5 is functionally active in both an ARE gene reporter assay and an Nrf2 nuclear translocation assay. The stereospecific nature of binding between (SRS)-5 and Keap1 as well as the preliminary but tractable structure-activity relationships support its use as a lead for our ongoing optimization.


Nature Chemical Biology | 2016

Correlating chemical sensitivity and basal gene expression reveals mechanism of action

Matthew G. Rees; Brinton Seashore-Ludlow; Jaime H. Cheah; Drew J. Adams; Edmund Price; Shubhroz Gill; Sarah Javaid; Matthew E. Coletti; Victor Victor Jones; Nicole E Bodycombe; Christian K. Soule; Benjamin Alexander; Ava Li; Philip Montgomery; Joanne Kotz; C. Suk-Yee Hon; Benito Munoz; Ted Liefeld; Vlado Dančík; Daniel A. Haber; Clary B. Clish; Joshua Bittker; Michelle Palmer; Bridget K. Wagner; Paul A. Clemons; Alykhan F. Shamji; Stuart L. Schreiber

Changes in cellular gene expression in response to small-molecule or genetic perturbations have yielded signatures that can connect unknown mechanisms of action (MoA) to ones previously established. We hypothesized that differential basal gene expression could be correlated with patterns of small-molecule sensitivity across many cell lines to illuminate the actions of compounds whose MoA are unknown. To test this idea, we correlated the sensitivity patterns of 481 compounds with ~19,000 basal transcript levels across 823 different human cancer cell lines and identified selective outlier transcripts. This process yielded many novel mechanistic insights, including the identification of activation mechanisms, cellular transporters, and direct protein targets. We found that ML239, originally identified in a phenotypic screen for selective cytotoxicity in breast cancer stem-like cells, most likely acts through activation of fatty acid desaturase 2 (FADS2). These data and analytical tools are available to the research community through the Cancer Therapeutics Response Portal.


Nature Chemical Biology | 2013

Niche-based screening identifies small-molecule inhibitors of leukemia stem cells

Kimberly A. Hartwell; Peter Miller; Siddhartha Mukherjee; Alissa R. Kahn; Alison L. Stewart; David J. Logan; Joseph Negri; Mildred Duvet; Marcus Järås; Rishi V. Puram; Vlado Dančík; Fatima Al-Shahrour; Thomas Kindler; Zuzana Tothova; Shrikanta Chattopadhyay; Thomas Hasaka; Rajiv Narayan; Mingji Dai; Christina Huang; Sebastian Shterental; Lisa P. Chu; J. Erika Haydu; Jae Hung Shieh; David P. Steensma; Benito Munoz; Joshua Bittker; Alykhan F. Shamji; Paul A. Clemons; Nicola Tolliday; Anne E. Carpenter

Efforts to develop more effective therapies for acute leukemia may benefit from high-throughput screening systems that reflect the complex physiology of the disease, including leukemia stem cells (LSCs) and supportive interactions with the bone marrow microenvironment. The therapeutic targeting of LSCs is challenging because LSCs are highly similar to normal hematopoietic stem and progenitor cells (HSPCs) and are protected by stromal cells in vivo. We screened 14,718 compounds in a leukemia-stroma co-culture system for inhibition of cobblestone formation, a cellular behavior associated with stem-cell function. Among those compounds that inhibited malignant cells but spared HSPCs was the cholesterol-lowering drug lovastatin. Lovastatin showed anti-LSC activity in vitro and in an in vivo bone marrow transplantation model. Mechanistic studies demonstrated that the effect was on target, via inhibition of HMG-CoA reductase. These results illustrate the power of merging physiologically relevant models with high-throughput screening.


Nature Biotechnology | 2010

Towards patient-based cancer therapeutics

Stuart L. Schreiber; Alykhan F. Shamji; Paul A. Clemons; Cindy Hon; Angela N. Koehler; Benito Munoz; Michelle Palmer; Bridget K. Wagner; Scott Powers; Scott W. Lowe; Xuecui Guo; Alexander Krasnitz; Eric T. Sawey; Raffaella Sordella; Lincoln Stein; Lloyd C. Trotman; Riccardo Dalla-Favera; Adolfo A. Ferrando; Antonio Iavarone; Laura Pasqualucci; Jose M. Silva; Brent R. Stockwell; William C. Hahn; Lynda Chin; Ronald A. DePinho; Jesse S. Boehm; Shuba Gopal; Alan Huang; David E. Root; Barbara A. Weir

A new approach to the discovery of cancer therapeutics is emerging that begins with the cancer patient. Genomic analysis of primary tumors is providing an unprecedented molecular characterization of the disease. The next step requires relating the genetic features of cancers to acquired gene and pathway dependencies and identifying small-molecule therapeutics that target them.


Nature | 2016

Diversity-oriented synthesis yields novel multistage antimalarial inhibitors

Nobutaka Kato; Eamon Comer; Tomoyo Sakata-Kato; Arvind Sharma; Manmohan Sharma; Micah Maetani; Jessica Bastien; Nicolas M. B. Brancucci; Joshua Bittker; Victoria C. Corey; David C. Clarke; Emily R. Derbyshire; Gillian L. Dornan; Sandra Duffy; Sean Eckley; Maurice A. Itoe; Karin M. J. Koolen; Timothy A. Lewis; Ping S. Lui; Amanda K Lukens; Emily Lund; Sandra March; Elamaran Meibalan; Bennett C. Meier; Jacob A. McPhail; Branko Mitasev; Eli L. Moss; Morgane Sayes; Yvonne Van Gessel; Mathias J. Wawer

Antimalarial drugs have thus far been chiefly derived from two sources—natural products and synthetic drug-like compounds. Here we investigate whether antimalarial agents with novel mechanisms of action could be discovered using a diverse collection of synthetic compounds that have three-dimensional features reminiscent of natural products and are underrepresented in typical screening collections. We report the identification of such compounds with both previously reported and undescribed mechanisms of action, including a series of bicyclic azetidines that inhibit a new antimalarial target, phenylalanyl-tRNA synthetase. These molecules are curative in mice at a single, low dose and show activity against all parasite life stages in multiple in vivo efficacy models. Our findings identify bicyclic azetidines with the potential to both cure and prevent transmission of the disease as well as protect at-risk populations with a single oral dose, highlighting the strength of diversity-oriented synthesis in revealing promising therapeutic targets.


ACS Medicinal Chemistry Letters | 2012

Diversity-Oriented Synthesis Yields a Novel Lead for the Treatment of Malaria

Richard Heidebrecht; Carol Mulrooney; Christopher P. Austin; Robert Barker; Jennifer A. Beaudoin; Ken Chih-Chien Cheng; Eamon Comer; Sivaraman Dandapani; Justin Dick; Jeremy R. Duvall; Eric Ekland; David A. Fidock; Mark E. Fitzgerald; Michael A. Foley; Rajarshi Guha; Paul L. Hinkson; Martin Kramer; Amanda K Lukens; Daniela Masi; Lisa A. Marcaurelle; Xin-Zhuan Su; Craig J. Thomas; Michel Weiwer; Roger Wiegand; Dyann F. Wirth; Menghang Xia; Jing Yuan; Jinghua Zhao; Michelle Palmer; Benito Munoz

Here, we describe the discovery of a novel antimalarial agent using phenotypic screening of Plasmodium falciparum asexual blood-stage parasites. Screening a novel compound collection created using diversity-oriented synthesis (DOS) led to the initial hit. Structure–activity relationships guided the synthesis of compounds having improved potency and water solubility, yielding a subnanomolar inhibitor of parasite asexual blood-stage growth. Optimized compound 27 has an excellent off-target activity profile in erythrocyte lysis and HepG2 assays and is stable in human plasma. This compound is available via the molecular libraries probe production centers network (MLPCN) and is designated ML238.


Bioorganic & Medicinal Chemistry Letters | 2012

Identification of a Selective Small Molecule Inhibitor of Breast Cancer Stem Cells

Andrew Germain; Leigh C. Carmody; Barbara Morgan; Cristina Fernandez; Erin Forbeck; Tim Lewis; Partha Nag; Amal Ting; Lynn VerPlank; Yuxiong Feng; Jose R. Perez; Sivaraman Dandapani; Michelle Palmer; Eric S. Lander; Piyush B. Gupta; Stuart L. Schreiber; Benito Munoz

A high-throughput screen (HTS) with the National Institute of Health-Molecular Libraries Small Molecule Repository (NIH-MLSMR) compound collection identified a class of acyl hydrazones to be selectively lethal to breast cancer stem cell (CSC) enriched populations. Medicinal chemistry efforts were undertaken to optimize potency and selectivity of this class of compounds. The optimized compound was declared as a probe (ML239) with the NIH Molecular Libraries Program and displayed greater than 20-fold selective inhibition of the breast CSC-like cell line (HMLE_sh_Ecad) over the isogenic control line (HMLE_sh_GFP).

Collaboration


Dive into the Benito Munoz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stuart L Schreiber

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin Vincent

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luke Whitesell

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge