Network


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

Hotspot


Dive into the research topics where Justin M. Roberts is active.

Publication


Featured researches published by Justin M. Roberts.


Science | 2015

Phthalimide conjugation as a strategy for in vivo target protein degradation

Georg E. Winter; Dennis L. Buckley; Joshiawa Paulk; Justin M. Roberts; Amanda Souza; Sirano Dhe-Paganon; James E. Bradner

A degrading game plan for cancer therapy Certain classes of proteins that contribute to cancer development are challenging to target therapeutically. Winter et al. devised a chemical strategy that, in principle, permits the selective degradation of any protein of interest. The strategy involves chemically attaching a ligand known to bind the desired protein to another molecule that hijacks an enzyme whose function is to direct proteins to the cells protein degradation machinery. In a proof-of-concept study, they demonstrated selective degradation of a transcriptional coactivator called bromodomain-containing protein 4 and delayed the progression of leukemia in mice. Science, this issue p. 1376 A chemical strategy that leads to selective destruction of proteins of interest may be a valuable tool for drug development. The development of effective pharmacological inhibitors of multidomain scaffold proteins, notably transcription factors, is a particularly challenging problem. In part, this is because many small-molecule antagonists disrupt the activity of only one domain in the target protein. We devised a chemical strategy that promotes ligand-dependent target protein degradation using as an example the transcriptional coactivator BRD4, a protein critical for cancer cell growth and survival. We appended a competitive antagonist of BET bromodomains to a phthalimide moiety to hijack the cereblon E3 ubiquitin ligase complex. The resultant compound, dBET1, induced highly selective cereblon-dependent BET protein degradation in vitro and in vivo and delayed leukemia progression in mice. A second series of probes resulted in selective degradation of the cytosolic protein FKBP12. This chemical strategy for controlling target protein stability may have implications for therapeutically targeting previously intractable proteins.


Science | 2015

DRUG DEVELOPMENT. Phthalimide conjugation as a strategy for in vivo target protein degradation.

Georg E. Winter; Dennis L. Buckley; Joshiawa Paulk; Justin M. Roberts; Amanda Souza; Sirano Dhe-Paganon; James E. Bradner

A degrading game plan for cancer therapy Certain classes of proteins that contribute to cancer development are challenging to target therapeutically. Winter et al. devised a chemical strategy that, in principle, permits the selective degradation of any protein of interest. The strategy involves chemically attaching a ligand known to bind the desired protein to another molecule that hijacks an enzyme whose function is to direct proteins to the cells protein degradation machinery. In a proof-of-concept study, they demonstrated selective degradation of a transcriptional coactivator called bromodomain-containing protein 4 and delayed the progression of leukemia in mice. Science, this issue p. 1376 A chemical strategy that leads to selective destruction of proteins of interest may be a valuable tool for drug development. The development of effective pharmacological inhibitors of multidomain scaffold proteins, notably transcription factors, is a particularly challenging problem. In part, this is because many small-molecule antagonists disrupt the activity of only one domain in the target protein. We devised a chemical strategy that promotes ligand-dependent target protein degradation using as an example the transcriptional coactivator BRD4, a protein critical for cancer cell growth and survival. We appended a competitive antagonist of BET bromodomains to a phthalimide moiety to hijack the cereblon E3 ubiquitin ligase complex. The resultant compound, dBET1, induced highly selective cereblon-dependent BET protein degradation in vitro and in vivo and delayed leukemia progression in mice. A second series of probes resulted in selective degradation of the cytosolic protein FKBP12. This chemical strategy for controlling target protein stability may have implications for therapeutically targeting previously intractable proteins.


Nature | 2017

Transcription control by the ENL YEATS domain in acute leukaemia

Michael A. Erb; Thomas G. Scott; Bin E. Li; Huafeng Xie; Joshiawa Paulk; Hyuk-Soo Seo; Amanda Souza; Justin M. Roberts; Shiva Dastjerdi; Dennis L. Buckley; Neville E. Sanjana; Ophir Shalem; Behnam Nabet; Rhamy Zeid; Nana K. Offei-Addo; Sirano Dhe-Paganon; Feng Zhang; Stuart H. Orkin; Georg E. Winter; James E. Bradner

Recurrent chromosomal translocations producing a chimaeric MLL oncogene give rise to a highly aggressive acute leukaemia associated with poor clinical outcome. The preferential involvement of chromatin-associated factors as MLL fusion partners belies a dependency on transcription control. Despite recent progress made in targeting chromatin regulators in cancer, available therapies for this well-characterized disease remain inadequate, prompting the need to identify new targets for therapeutic intervention. Here, using unbiased CRISPR–Cas9 technology to perform a genome-scale loss-of-function screen in an MLL-AF4-positive acute leukaemia cell line, we identify ENL as an unrecognized gene that is specifically required for proliferation in vitro and in vivo. To explain the mechanistic role of ENL in leukaemia pathogenesis and dynamic transcription control, a chemical genetic strategy was developed to achieve targeted protein degradation. Acute loss of ENL suppressed the initiation and elongation of RNA polymerase II at active genes genome-wide, with pronounced effects at genes featuring a disproportionate ENL load. Notably, an intact YEATS chromatin-reader domain was essential for ENL-dependent leukaemic growth. Overall, these findings identify a dependency factor in acute leukaemia and suggest a mechanistic rationale for disrupting the YEATS domain in disease.


Nature Chemical Biology | 2016

Design and characterization of bivalent BET inhibitors

Minoru Tanaka; Justin M. Roberts; Hyuk-Soo Seo; Amanda Souza; Joshiawa Paulk; Thomas G. Scott; Stephen L. DeAngelo; Sirano Dhe-Paganon; James E. Bradner

Cellular signaling is often propagated by multivalent interactions. Multivalency creates avidity, allowing stable biophysical recognition. Multivalency is an attractive strategy for achieving potent binding to protein targets, as the affinity of bivalent ligands is often greater than the sum of monovalent affinities. The BET family of transcriptional coactivators features tandem bromodomains, through which BET proteins naturally bind acetylated histones and transcription factors. All reported BRD4 antagonists bind in a monovalent fashion. Here, we report the first bivalent BET bromodomain inhibitor, MT1 that has unprecedented potency. Biophysical and biochemical studies suggest MT1 is an intramolecular bivalent BRD4 binder that is over 100-fold more potent in cellular assays compared to the corresponding monovalent antagonist, JQ1. MT1 significantly delayed leukemia progression in mice (Mus musculus) compared to JQ1. These data qualify a powerful chemical probe for BET bromodomains and extensible rationale for further development of multidomain epigenetic reader protein inhibitors.


Pharmaceutical patent analyst | 2015

Inhibitors of emerging epigenetic targets for cancer therapy: a patent review (2010-2014).

Minoru Tanaka; Justin M. Roberts; Jun Qi; James E. Bradner

Gene regulatory pathways comprise an emerging and active area of chemical probe discovery and investigational drug development. Emerging insights from cancer genome sequencing and chromatin biology have identified leveraged opportunities for development of chromatin-directed small molecules as cancer therapies. At present, only six agents in two epigenetic target classes have been approved by the US FDA, limited to treatment of hematological malignancies. Recently, new classes of epigenetic inhibitors have appeared in literatures. First-in-class compounds have successfully transitioned to clinical investigation, importantly also in solid tumors and pediatric malignancies. This review considers patent applications for small-molecule inhibitors of selected epigenetic targets from 2010 to 2014. Included are exemplary classes of chromatin-associated epigenomic writers (DOT1L and EZH2), erasers (LSD1) and readers (BRD4).


Nature Communications | 2018

Targeting the CoREST complex with dual histone deacetylase and demethylase inhibitors

Jay H. Kalin; Muzhou Wu; Andrea V. Gómez; Yun Song; Jayanta Das; Dawn Hayward; Nkosi Adejola; Mingxuan Wu; Izabela P. Panova; Hye Jin Chung; Edward Kim; Holly J. Roberts; Justin M. Roberts; Polina Prusevich; Jeliazko R. Jeliazkov; Shourya S. Roy Burman; Louise Fairall; Charles P. Milano; Abdulkerim Eroglu; Charlotte M. Proby; Albena T. Dinkova-Kostova; Wayne W. Hancock; Jeffrey J. Gray; James E. Bradner; Sergio Valente; Antonello Mai; Nicole M. Anders; Michelle A. Rudek; Yong Hu; Byungwoo Ryu

Here we report corin, a synthetic hybrid agent derived from the class I HDAC inhibitor (entinostat) and an LSD1 inhibitor (tranylcypromine analog). Enzymologic analysis reveals that corin potently targets the CoREST complex and shows more sustained inhibition of CoREST complex HDAC activity compared with entinostat. Cell-based experiments demonstrate that corin exhibits a superior anti-proliferative profile against several melanoma lines and cutaneous squamous cell carcinoma lines compared to its parent monofunctional inhibitors but is less toxic to melanocytes and keratinocytes. CoREST knockdown, gene expression, and ChIP studies suggest that corin’s favorable pharmacologic effects may rely on an intact CoREST complex. Corin was also effective in slowing tumor growth in a melanoma mouse xenograft model. These studies highlight the promise of a new class of two-pronged hybrid agents that may show preferential targeting of particular epigenetic regulatory complexes and offer unique therapeutic opportunities.Alteration of the epigenetic landscape has been implicated in several disease processes, where targeting histone modifiers may have therapeutic applications. Here the authors report a bifunctional small molecule inhibitor that simultaneously targets the deacetylase (HDAC1) and demethylase (LSD1) activities of the CoREST complex.


eLife | 2017

MELK is not necessary for the proliferation of basal-like breast cancer cells

Hai-Tsang Huang; Hyuk-Soo Seo; Tinghu Zhang; Yubao Wang; Baishan Jiang; Qing Li; Dennis L. Buckley; Behnam Nabet; Justin M. Roberts; Joshiawa Paulk; Shiva Dastjerdi; Georg E. Winter; Hilary McLauchlan; Jennifer Moran; James E. Bradner; Michael J. Eck; Sirano Dhe-Paganon; Jean Zhao; Nathanael S. Gray

Thorough preclinical target validation is essential for the success of drug discovery efforts. In this study, we combined chemical and genetic perturbants, including the development of a novel selective maternal embryonic leucine zipper kinase (MELK) inhibitor HTH-01-091, CRISPR/Cas9-mediated MELK knockout, a novel chemical-induced protein degradation strategy, RNA interference and CRISPR interference to validate MELK as a therapeutic target in basal-like breast cancers (BBC). In common culture conditions, we found that small molecule inhibition, genetic deletion, or acute depletion of MELK did not significantly affect cellular growth. This discrepancy to previous findings illuminated selectivity issues of the widely used MELK inhibitor OTSSP167, and potential off-target effects of MELK-targeting short hairpins. The different genetic and chemical tools developed here allow for the identification and validation of any causal roles MELK may play in cancer biology, which will be required to guide future MELK drug discovery efforts. Furthermore, our study provides a general framework for preclinical target validation.


Current protocols in chemical biology | 2015

A Bead‐Based Proximity Assay for BRD4 Ligand Discovery

Justin M. Roberts; James E. Bradner

Bromodomain‐containing proteins have emerged as desirable targets for anti‐neoplastic and anti‐inflammatory drug discovery. Toward the development of selective inhibitors of the BET family of bromodomains, we optimized bead‐based assays to detect interactions between bromodomains and poly‐acetylated histone peptides. Donor and acceptor beads bound to target and ligand are brought into proximity by this protein‐protein interaction. After laser illumination, singlet oxygen evolved from donor beads travels to the spatially close acceptor beads, resulting in chemiluminesence. This AlphaScreen assay has proven amendable to high‐throughput screening, secondary validation, and specificity profiling during lead discovery and optimization. Here we report our protocol for assay development to measure inhibition of ligand binding to bromodomain‐containing protein 4 (BRD4). We discuss the discovery of an appropriate probe, optimization of bead, probe, and protein concentrations, and the derivation of protein‐probe inhibition curves. Finally, we explore the implementation of this technology for high‐throughput screening of potential BRD4 inhibitors.


Nature Chemical Biology | 2018

The dTAG system for immediate and target-specific protein degradation

Behnam Nabet; Justin M. Roberts; Dennis L. Buckley; Joshiawa Paulk; Shiva Dastjerdi; Annan Yang; Alan L. Leggett; Michael A. Erb; Matthew A. Lawlor; Amanda Souza; Thomas G. Scott; Sarah Vittori; Jennifer A. Perry; Jun Qi; Georg E. Winter; Kwok-Kin Wong; Nathanael S. Gray; James E. Bradner

Dissection of complex biological systems requires target-specific control of the function or abundance of proteins. Genetic perturbations are limited by off-target effects, multicomponent complexity, and irreversibility. Most limiting is the requisite delay between modulation to experimental measurement. To enable the immediate and selective control of single protein abundance, we created a chemical biology system that leverages the potency of cell-permeable heterobifunctional degraders. The dTAG system pairs a novel degrader of FKBP12F36V with expression of FKBP12F36V in-frame with a protein of interest. By transgene expression or CRISPR-mediated locus-specific knock-in, we exemplify a generalizable strategy to study the immediate consequence of protein loss. Using dTAG, we observe an unexpected superior antiproliferative effect of pan-BET bromodomain degradation over selective BRD4 degradation, characterize immediate effects of KRASG12V loss on proteomic signaling, and demonstrate rapid degradation in vivo. This technology platform will confer kinetic resolution to biological investigation and provide target validation in the context of drug discovery.The dTAG system pairs potent heterobifunctional degraders and extensible tagging strategies to achieve immediate and reversible degradation of divergent proteins, facilitating biological investigation and drug target validation in cells and in mice.


ChemMedChem | 2016

Assessment of Bromodomain Target Engagement by a Series of BI2536 Analogues with Miniaturized BET-BRET

Luke W. Koblan; Dennis L. Buckley; Christopher J. Ott; Mark E. Fitzgerald; Stuart W. J. Ember; Jin-Yi Zhu; Shuai Liu; Justin M. Roberts; David Remillard; Sarah Vittori; Wei Zhang; Ernst Schönbrunn; James E. Bradner

Evaluating the engagement of a small molecule ligand with a protein target in cells provides useful information for chemical probe optimization and pharmaceutical development. While several techniques exist that can be performed in a low‐throughput manner, systematic evaluation of large compound libraries remains a challenge. In‐cell engagement measurements are especially useful when evaluating compound classes suspected to target multiple cellular factors. In this study we used a bioluminescent resonant energy transfer assay to assess bromodomain engagement by a compound series containing bromodomain‐ and kinase‐biasing polypharmacophores based on the known dual BRD4 bromodomain/PLK1 kinase inhibitor BI2536. With this assay, we discovered several novel agents with bromodomain‐selective specificity profiles and cellular activity. Thus, this platform aids in distinguishing molecules whose cellular activity is difficult to assess due to polypharmacologic effects.

Collaboration


Dive into the Justin M. Roberts'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

Behnam Nabet

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge