Network


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

Hotspot


Dive into the research topics where Lesley A. Mathews Griner is active.

Publication


Featured researches published by Lesley A. Mathews Griner.


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.


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

Blockade of oncogenic IκB kinase activity in diffuse large B-cell lymphoma by bromodomain and extraterminal domain protein inhibitors

Michele Ceribelli; Priscilla N. Kelly; Arthur L. Shaffer; George W. Wright; Wenming Xiao; Yibin Yang; Lesley A. Mathews Griner; Rajarshi Guha; Paul Shinn; Jonathan M. Keller; Dongbo Liu; Paresma R. Patel; Marc Ferrer; Shivangi Joshi; Sujata Nerle; Peter Sandy; Emmanuel Normant; Craig J. Thomas; Louis M. Staudt

Significance The activated B-cell–like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) is an aggressive cancer that can only be cured in roughly 40% of cases. These malignant cells rely on the NF-κB signaling pathway for survival. Here, we report that genetic or pharmacologic interference with bromodomain and extraterminal domain (BET) chromatin proteins reduces NF-κB activity and ABC DLBCL viability. Unexpectedly, the mechanism involves inhibition of IκB kinase, the key cytoplasmic enzyme that activates the NF-κB pathway. The NF-κB pathway in ABC DLBCL is activated by B-cell receptor signaling, which can be blocked by the BTK kinase inhibitor ibrutinib. BET inhibitors synergized with ibrutinib to decrease growth of ABC DLBCL tumors in mouse models. BET inhibitors should be evaluated in ABC DLBCL clinical trials. In the activated B-cell–like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL), NF-κB activity is essential for viability of the malignant cells and is sustained by constitutive activity of IκB kinase (IKK) in the cytoplasm. Here, we report an unexpected role for the bromodomain and extraterminal domain (BET) proteins BRD2 and BRD4 in maintaining oncogenic IKK activity in ABC DLBCL. IKK activity was reduced by small molecules targeting BET proteins as well as by genetic knockdown of BRD2 and BRD4 expression, thereby inhibiting downstream NF-κB–driven transcriptional programs and killing ABC DLBCL cells. Using a high-throughput platform to screen for drug–drug synergy, we observed that the BET inhibitor JQ1 combined favorably with multiple drugs targeting B-cell receptor signaling, one pathway that activates IKK in ABC DLBCL. The BTK kinase inhibitor ibrutinib, which is in clinical development for the treatment of ABC DLBCL, synergized strongly with BET inhibitors in killing ABC DLBCL cells in vitro and in a xenograft mouse model. These findings provide a mechanistic basis for the clinical development of BET protein inhibitors in ABC DLBCL, particularly in combination with other modulators of oncogenic IKK signaling.


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.


Oncotarget | 2015

Aurora B kinase is a potent and selective target in MYCN-driven neuroblastoma

Dominik Bogen; Jun S. Wei; David O. Azorsa; Pinar Ormanoglu; Eugen Buehler; Rajarshi Guha; Jonathan M. Keller; Lesley A. Mathews Griner; Marc Ferrer; Young K. Song; Hongling Liao; Arnulfo Mendoza; Berkley Gryder; Sivasish Sindri; Jianbin He; Xinyu Wen; Shile Zhang; John F. Shern; Marielle E. Yohe; Sabine Taschner-Mandl; Jason M. Shohet; Craig J. Thomas; Scott E. Martin; Peter F. Ambros; Javed Khan

Despite advances in multimodal treatment, neuroblastoma (NB) is often fatal for children with high-risk disease and many survivors need to cope with long-term side effects from high-dose chemotherapy and radiation. To identify new therapeutic targets, we performed an siRNA screen of the druggable genome combined with a small molecule screen of 465 compounds targeting 39 different mechanisms of actions in four NB cell lines. We identified 58 genes as targets, including AURKB, in at least one cell line. In the drug screen, aurora kinase inhibitors (nine molecules) and in particular the AURKB-selective compound, barasertib, were the most discriminatory with regard to sensitivity for MYCN-amplified cell lines. In an expanded panel of ten NB cell lines, those with MYCN-amplification and wild-type TP53 were the most sensitive to low nanomolar concentrations of barasertib. Inhibition of the AURKB kinase activity resulted in decreased phosphorylation of the known target, histone H3, and upregulation of TP53 in MYCN-amplified, TP53 wild-type cells. However, both wild-type and TP53 mutant MYCN-amplified cell lines arrested in G2/M phase upon AURKB inhibition. Additionally, barasertib induced endoreduplication and apoptosis. Treatment of MYCN-amplified/TP53 wild-type neuroblastoma xenografts resulted in profound growth inhibition and tumor regression. Therefore, aurora B kinase inhibition is highly effective in aggressive neuroblastoma and warrants further investigation in clinical trials.


PLOS ONE | 2014

A High Throughput Screening Assay System for the Identification of Small Molecule Inhibitors of gsp

Nisan Bhattacharyya; Xin Hu; Catherine Z. Chen; Lesley A. Mathews Griner; Wei Zheng; James Inglese; Christopher P. Austin; Juan J. Marugan; Noel Southall; Susanne Neumann; John K. Northup; Marc Ferrer; Michael T. Collins

Mis-sense mutations in the α-subunit of the G-protein, Gsα, cause fibrous dysplasia of bone/McCune-Albright syndrome. The biochemical outcome of these mutations is constitutively active Gsα and increased levels of cAMP. The aim of this study was to develop an assay system that would allow the identification of small molecule inhibitors specific for the mutant Gsα protein, the so-called gsp oncogene. Commercially available Chinese hamster ovary cells were stably transfected with either wild-type (WT) or mutant Gsα proteins (R201C and R201H). Stable cell lines with equivalent transfected Gsα protein expression that had relatively lower (WT) or higher (R201C and R201H) cAMP levels were generated. These cell lines were used to develop a fluorescence resonance energy transfer (FRET)–based cAMP assay in 1536-well microplate format for high throughput screening of small molecule libraries. A small molecule library of 343,768 compounds was screened to identify modulators of gsp activity. A total of 1,356 compounds with inhibitory activity were initially identified and reconfirmed when tested in concentration dose responses. Six hundred eighty-six molecules were selected for further analysis after removing cytotoxic compounds and those that were active in forskolin-induced WT cells. These molecules were grouped by potency, efficacy, and structural similarities to yield 22 clusters with more than 5 of structurally similar members and 144 singleton molecules. Seven chemotypes of the major clusters were identified for further testing and analyses.


Journal of Biomolecular Screening | 2015

Small Molecule, NSC95397, Inhibits the CtBP1-Protein Partner Interaction and CtBP1-Mediated Transcriptional Repression

Melanie A. Blevins; Jennifer Kouznetsova; Aaron B. Krueger; Rebecca King; Lesley A. Mathews Griner; Xin Hu; Noel Southall; Juan J. Marugan; Qinghong Zhang; Marc Ferrer; Rui Zhao

Carboxyl-terminal binding protein (CtBP) is a transcriptional corepressor that suppresses multiple proapoptotic and epithelial genes. CtBP is overexpressed in many human cancers, and its overexpression increases stem cell–like features, epithelial-mesenchymal transition, and cancer cell survival. Knockdown of CtBP also increases apoptosis independent of p53 in cell culture. Therefore, targeting CtBP with small molecules that disrupt its interaction with transcription factor partners may be an effective cancer therapy. To elicit its corepressing effect, CtBP binds to a conserved peptide motif in each transcription factor partner. We developed an AlphaScreen high-throughput screening assay to monitor the interaction between CtBP and E1A (which mimics the interaction between CtBP and its transcriptional partners). We screened the LOPAC library of 1280 bioactive compounds and identified NSC95397, which inhibits the CtBP-E1A interaction (IC50 = 2.9 µM). The inhibitory activity of NSC95397 was confirmed using two secondary assays and a counterscreen. NSC95397 also behaved as a weak substrate of CtBP dehydrogenase activity and did not inhibit another dehydrogenase, lactase dehydrogenase. Finally, NSC95397 was able to disrupt CtBP-mediated transcriptional repression of a target gene. These studies present a new possibility for the development of a therapeutic agent targeting tumors through disrupting the CtBP transcriptional complex.


PLOS Pathogens | 2016

Efficacy and Mechanism of Action of Low Dose Emetine against Human Cytomegalovirus

Rupkatha Mukhopadhyay; Sujayita Roy; Rajkumar Venkatadri; Yu Pin Su; Wenjuan Ye; Elena Barnaeva; Lesley A. Mathews Griner; Noel Southall; Xin Hu; Amy Wang; Xin Xu; Andrés E. Dulcey; Juan J. Marugan; Marc Ferrer; Ravit Arav-Boger

Infection with human cytomegalovirus (HCMV) is a threat for pregnant women and immunocompromised hosts. Although limited drugs are available, development of new agents against HCMV is desired. Through screening of the LOPAC library, we identified emetine as HCMV inhibitor. Additional studies confirmed its anti-HCMV activities in human foreskin fibroblasts: EC50−40±1.72 nM, CC50−8±0.56 μM, and selectivity index of 200. HCMV inhibition occurred after virus entry, but before DNA replication, and resulted in decreased expression of viral proteins. Synergistic virus inhibition was achieved when emetine was combined with ganciclovir. In a mouse CMV (MCMV) model, emetine was well-tolerated, displayed long half-life, preferential distribution to tissues over plasma, and effectively suppressed MCMV. Since the in vitro anti-HCMV activity of emetine decreased significantly in low-density cells, a mechanism involving cell cycle regulation was suspected. HCMV inhibition by emetine depended on ribosomal processing S14 (RPS14) binding to MDM2, leading to disruption of HCMV-induced MDM2-p53 and MDM2-IE2 interactions. Irrespective of cell density, emetine induced RPS14 translocation into the nucleus during infection. In infected high-density cells, MDM2 was available for interaction with RPS14, resulting in disruption of MDM2-p53 interaction. However, in low-density cells the pre-existing interaction of MDM2-p53 could not be disrupted, and RPS14 could not interact with MDM2. In high-density cells the interaction of MDM2-RPS14 resulted in ubiquitination and degradation of RPS14, which was not observed in low-density cells. In infected-only or in non-infected emetine-treated cells, RPS14 failed to translocate into the nucleus, hence could not interact with MDM2, and was not ubiquitinated. HCMV replicated similarly in RPS14 knockdown or control cells, but emetine did not inhibit virus replication in the former cell line. The interaction of MDM2-p53 was maintained in infected RPS14 knockdown cells despite emetine treatment, confirming a unique mechanism by which emetine exploits RPS14 to disrupt MDM2-p53 interaction. Summarized, emetine may represent a promising candidate for HCMV therapy alone or in combination with ganciclovir through a novel host-dependent mechanism.


Cell Death and Disease | 2016

Large-scale pharmacological profiling of 3D tumor models of cancer cells

Lesley A. Mathews Griner; Xiaohu Zhang; Rajarshi Guha; Crystal McKnight; Ian S. Goldlust; Madhu Lal-Nag; Kelli Wilson; Sam Michael; Steve Titus; Paul Shinn; Craig J. Thomas; Marc Ferrer

The discovery of chemotherapeutic agents for the treatment of cancer commonly uses cell proliferation assays in which cells grow as two-dimensional (2D) monolayers. Compounds identified using 2D monolayer assays often fail to advance during clinical development, most likely because these assays do not reproduce the cellular complexity of tumors and their microenvironment in vivo. The use of three-dimensional (3D) cellular systems have been explored as enabling more predictive in vitro tumor models for drug discovery. To date, small-scale screens have demonstrated that pharmacological responses tend to differ between 2D and 3D cancer cell growth models. However, the limited scope of screens using 3D models has not provided a clear delineation of the cellular pathways and processes that differentially regulate cell survival and death in the different in vitro tumor models. Here we sought to further understand the differences in pharmacological responses between cancer tumor cells grown in different conditions by profiling a large collection of 1912 chemotherapeutic agents. We compared pharmacological responses obtained from cells cultured in traditional 2D monolayer conditions with those responses obtained from cells forming spheres versus cells already in 3D spheres. The target annotation of the compound library screened enabled the identification of those key cellular pathways and processes that when modulated by drugs induced cell death in all growth conditions or selectively in the different cell growth models. In addition, we also show that many of the compounds targeting these key cellular functions can be combined to produce synergistic cytotoxic effects, which in many cases differ in the magnitude of their synergism depending on the cellular model and cell type. The results from this work provide a high-throughput screening framework to profile the responses of drugs both as single agents and in pairwise combinations in 3D sphere models of cancer cells.


Science Translational Medicine | 2018

Metarrestin, a perinucleolar compartment inhibitor, effectively suppresses metastasis

Kevin J. Frankowski; Chen Wang; Samarjit Patnaik; Frank J. Schoenen; Noel Southall; Dandan Li; Yaroslav Teper; Wei Sun; Irawati Kandela; Deqing Hu; Christopher Dextras; Zachary Knotts; Yansong Bian; John Norton; Steve Titus; Marzena A. Lewandowska; Yiping Wen; Katherine I. Farley; Lesley A. Mathews Griner; Jamey Sultan; Zhaojing Meng; Ming Zhou; Tomas Vilimas; Astin S. Powers; Serguei Kozlov; Kunio Nagashima; Humair S. Quadri; Min Fang; Charles Long; Ojus Khanolkar

A compound that reduces the prevalence of perinucleolar compartment in cancer cells inhibits metastasis in vivo. Blocking metastasis from the inside Metastasis, the spread of tumor cells that often results in cancer patients’ deaths, remains difficult to treat for all cancer types. To intervene with this process, Frankowski et al. searched for inhibitors of the perinucleolar compartment, a structure located within the nuclei of cancer cells and associated with metastatic capacity. Through high-throughput screening followed by chemical optimization, the authors developed a compound they called metarrestin, which disrupts the perinuclear compartment in different types of cancer cells. They discovered that metarrestin inhibited tumor invasion and metastasis in multiple mouse models of cancer and prolonged the animals’ survival, suggesting its potential relevance for translation to patients. Metastasis remains a leading cause of cancer mortality due to the lack of specific inhibitors against this complex process. To identify compounds selectively targeting the metastatic state, we used the perinucleolar compartment (PNC), a complex nuclear structure associated with metastatic behaviors of cancer cells, as a phenotypic marker for a high-content screen of over 140,000 structurally diverse compounds. Metarrestin, obtained through optimization of a screening hit, disassembles PNCs in multiple cancer cell lines, inhibits invasion in vitro, suppresses metastatic development in three mouse models of human cancer, and extends survival of mice in a metastatic pancreatic cancer xenograft model with no organ toxicity or discernable adverse effects. Metarrestin disrupts the nucleolar structure and inhibits RNA polymerase (Pol) I transcription, at least in part by interacting with the translation elongation factor eEF1A2. Thus, metarrestin represents a potential therapeutic approach for the treatment of metastatic cancer.


PLOS ONE | 2016

Chemical Screens Identify Drugs that Enhance or Mitigate Cellular Responses to Antibody-Toxin Fusion Proteins

Antonella Antignani; Lesley A. Mathews Griner; Rajarshi Guha; Nathan Simon; Matteo Pasetto; Jonathan M. Keller; Manjie Huang; Evan Angelus; Ira Pastan; Marc Ferrer; David J. FitzGerald; Craig J. Thomas

The intersection of small molecular weight drugs and antibody-based therapeutics is rarely studied in large scale. Both types of agents are currently part of the cancer armamentarium. However, very little is known about how to combine them in optimal ways. Immunotoxins are antibody-toxin gene fusion proteins engineered to target cancer cells via antibody binding to surface antigens. For fusion proteins derived from Pseudomonas exotoxin (PE), potency relies on the enzymatic domain of the toxin which catalyzes the ADP-ribosylation of EF2 causing inhibition of protein synthesis leading to cell death. Candidate immunotoxins have demonstrated clear value in clinical trials but generally have not been curative as single agents. Therefore we undertook three screens to discover effective combinations that could act synergistically. From the MIPE-3 library of compounds we identified various enhancers of immunotoxin action and at least one major class of inhibitor. Follow-up experiments confirmed the screening data and suggested that immunotoxins when administered with everolimus or nilotinib exhibit favorable combinatory activity and would be candidates for preclinical development. Mechanistic studies revealed that everolimus-immunotoxin combinations acted synergistically on elements of the protein synthetic machinery, including S61 kinase and 4E-BP1 of the mTORC1 pathway. Conversely, PARP inhibitors antagonized immunotoxins and also blocked the toxicity due to native ADP-ribosylating toxins. Thus, our goal of investigating a chemical library was justified based on the identification of several approved compounds that could be developed preclinically as ‘enhancers’ and at least one class of mitigator to be avoided.

Collaboration


Dive into the Lesley A. Mathews Griner's collaboration.

Top Co-Authors

Avatar

Marc Ferrer

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

Rajarshi Guha

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jonathan M. Keller

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Noel Southall

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paul Shinn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Juan J. Marugan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xin Hu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Damien Y. Duveau

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Scott E. Martin

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge