Berkley Gryder
National Institutes of Health
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Berkley Gryder.
Nature Communications | 2016
Elad Jacoby; Sang M. Nguyen; Thomas J. Fountaine; Kathryn Welp; Berkley Gryder; Haiying Qin; Yinmeng Yang; Christopher D. Chien; Alix E. Seif; Haiyan Lei; Young K. Song; Javed Khan; Daniel W. Lee; Crystal L. Mackall; Rebecca A. Gardner; Michael C. Jensen; Jack F. Shern; Terry J. Fry
Adoptive immunotherapy using chimeric antigen receptor (CAR) expressing T cells targeting the CD19 B lineage receptor has demonstrated marked success in relapsed pre-B-cell acute lymphoblastic leukaemia (ALL). Persisting CAR-T cells generate sustained pressure against CD19 that may drive unique mechanisms of resistance. Pre-B ALL originates from a committed pre-B cell or an earlier progenitor, with potential to reprogram into other hematopoietic lineages. Here we report changes in lineage markers including myeloid conversion in patients following CD19 CAR therapy. Using murine ALL models we study the long-term effects of CD19 CAR-T cells and demonstrate partial or complete lineage switch as a consistent mechanism of CAR resistance depending on the underlying genetic oncogenic driver. Deletion of Pax5 or Ebf1 recapitulates lineage reprogramming occurring during CD19 CAR pressure. Our findings establish lineage switch as a mechanism of CAR resistance exposing inherent plasticity in genetic subtypes of pre-B-cell ALL.
Clinical Cancer Research | 2016
Wendy Chang; Andrew S. Brohl; Rajesh Patidar; Sivasish Sindiri; Jack F. Shern; Jun S. Wei; Young K. Song; Marielle E. Yohe; Berkley Gryder; Shile Zhang; Kathleen A. Calzone; Nityashree Shivaprasad; Xinyu Wen; Thomas C. Badgett; Markku Miettinen; Kip R. Hartman; James C. League-Pascual; Toby Trahair; Brigitte C. Widemann; Melinda S. Merchant; Rosandra N. Kaplan; Jimmy Lin; Javed Khan
Purpose: We undertook a multidimensional clinical genomics study of children and adolescent young adults with relapsed and refractory cancers to determine the feasibility of genome-guided precision therapy. Experimental Design: Patients with non-central nervous system solid tumors underwent a combination of whole exome sequencing (WES), whole transcriptome sequencing (WTS), and high-density single-nucleotide polymorphism array analysis of the tumor, with WES of matched germline DNA. Clinically actionable alterations were identified as a reportable germline mutation, a diagnosis change, or a somatic event (including a single nucleotide variant, an indel, an amplification, a deletion, or a fusion gene), which could be targeted with drugs in existing clinical trials or with FDA-approved drugs. Results: Fifty-nine patients in 20 diagnostic categories were enrolled from 2010 to 2014. Ages ranged from 7 months to 25 years old. Seventy-three percent of the patients had prior chemotherapy, and the tumors from these patients with relapsed or refractory cancers had a higher mutational burden than that reported in the literature. Thirty patients (51% of total) had clinically actionable mutations, of which 24 (41%) had a mutation that was currently targetable in a clinical trial setting, 4 patients (7%) had a change in diagnosis, and 7 patients (12%) had a reportable germline mutation. Conclusions: We found a remarkably high number of clinically actionable mutations in 51% of the patients, and 12% with significant germline mutations. We demonstrated the clinical feasibility of next-generation sequencing in a diverse population of relapsed and refractory pediatric solid tumors. Clin Cancer Res; 22(15); 3810–20. ©2016 AACR.
Oncotarget | 2015
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.
eLife | 2017
Inês M. Tenente; Madeline Hayes; Myron S. Ignatius; Karin M. McCarthy; Marielle Yohe; Sivasish Sindiri; Berkley Gryder; Mariana L. Oliveira; Ashwin Ramakrishnan; Qin Tang; Eleanor Chen; G. Petur Nielsen; Javed Khan; David M. Langenau
Rhabdomyosarcoma (RMS) is a pediatric malignacy of muscle with myogenic regulatory transcription factors MYOD and MYF5 being expressed in this disease. Consensus in the field has been that expression of these factors likely reflects the target cell of transformation rather than being required for continued tumor growth. Here, we used a transgenic zebrafish model to show that Myf5 is sufficient to confer tumor-propagating potential to RMS cells and caused tumors to initiate earlier and have higher penetrance. Analysis of human RMS revealed that MYF5 and MYOD are mutually-exclusively expressed and each is required for sustained tumor growth. ChIP-seq and mechanistic studies in human RMS uncovered that MYF5 and MYOD bind common DNA regulatory elements to alter transcription of genes that regulate muscle development and cell cycle progression. Our data support unappreciated and dominant oncogenic roles for MYF5 and MYOD convergence on common transcriptional targets to regulate human RMS growth. DOI: http://dx.doi.org/10.7554/eLife.19214.001
Cell Reports | 2017
Xiao Ling Li; Murugan Subramanian; Matthew F. Jones; Ritu Chaudhary; Deepak K. Singh; Xinying Zong; Berkley Gryder; Sivasish Sindri; Min Mo; Aaron J. Schetter; Xinyu Wen; Swetha Parvathaneni; Dickran Kazandjian; Lisa M. Miller Jenkins; Wei Tang; Fathi Elloumi; Jennifer L. Martindale; Maite Huarte; Yuelin Zhu; Ana I. Robles; Susan M. Frier; Frank Rigo; Maggie Cam; Stefan Ambs; Sudha Sharma; Curtis C. Harris; Mary Dasso; Kannanganattu V. Prasanth; Ashish Lal
Basal p53 levels are tightly suppressed under normal conditions. Disrupting this regulation results in elevated p53 levels to induce cell cycle arrest, apoptosis, and tumor suppression. Here, we report the suppression of basal p53 levels by a nuclear, p53-regulated long noncoding RNA that we termed PURPL (p53 upregulated regulator of p53 levels). Targeted depletion of PURPL in colorectal cancer cells results in elevated basal p53 levels and induces growth defects in cell culture and in mouse xenografts. PURPL associates with MYBBP1A, a protein that binds to and stabilizes p53, and inhibits the formation of the p53-MYBBP1A complex. In the absence of PURPL, MYBBP1A interacts with and stabilizes p53. Silencing MYBBP1A significantly rescues basal p53 levels and proliferation in PURPL-deficient cells, suggesting that MYBBP1A mediates the effect of PURPL in regulating p53. These results reveal a p53-PURPL auto-regulatory feedback loop and demonstrate a role for PURPL in maintaining basal p53 levels.
Cancer Cell | 2017
Veronica Veschi; Zhihui Liu; Ty C. Voss; Laurent Ozbun; Berkley Gryder; Chunhua Yan; Ying Hu; Anqi Ma; Jian Jin; Sharlyn J. Mazur; Norris Lam; Bárbara Kunzler Souza; Giuseppe Giannini; Gordon L. Hager; C.H. Arrowsmith; Javed Khan; Ettore Appella; Carol J. Thiele
Given the paucity of druggable mutations in high-risk neuroblastoma (NB), we undertook chromatin-focused small interfering RNA and chemical screens to uncover epigenetic regulators critical for the differentiation block in high-risk NB. High-content Opera imaging identified 53 genes whose loss of expression led to a decrease in NB cell proliferation and 16 also induced differentiation. From these, the secondary chemical screen identified SETD8, the H4K20me1 methyltransferase, as a druggable NB target. Functional studies revealed that SETD8 ablation rescued the pro-apoptotic and cell-cycle arrest functions of p53 by decreasing p53K382me1, leading to activation of the p53 canonical pathway. In pre-clinical xenograft NB models, genetic or pharmacological (UNC0379) SETD8 inhibition conferred a significant survival advantage, providing evidence for SETD8 as a therapeutic target in NB.
eLife | 2017
Ritu Chaudhary; Berkley Gryder; Wendy S. Woods; Murugan Subramanian; Matthew F. Jones; Xiao Ling Li; Lisa M. Miller Jenkins; Svetlana A. Shabalina; Min Mo; Mary Dasso; Yu-an Yang; Lalage M. Wakefield; Yuelin Zhu; Susan M. Frier; Branden S. Moriarity; Kannanganattu V. Prasanth; Pablo Perez-Pinera; Ashish Lal
Thousands of long noncoding RNAs (lncRNAs) have been discovered, yet the function of the vast majority remains unclear. Here, we show that a p53-regulated lncRNA which we named PINCR (p53-induced noncoding RNA), is induced ~100-fold after DNA damage and exerts a prosurvival function in human colorectal cancer cells (CRC) in vitro and tumor growth in vivo. Targeted deletion of PINCR in CRC cells significantly impaired G1 arrest and induced hypersensitivity to chemotherapeutic drugs. PINCR regulates the induction of a subset of p53 targets involved in G1 arrest and apoptosis, including BTG2, RRM2B and GPX1. Using a novel RNA pulldown approach that utilized endogenous S1-tagged PINCR, we show that PINCR associates with the enhancer region of these genes by binding to RNA-binding protein Matrin 3 that, in turn, associates with p53. Our findings uncover a critical prosurvival function of a p53/PINCR/Matrin 3 axis in response to DNA damage in CRC cells. DOI: http://dx.doi.org/10.7554/eLife.23244.001
Journal of Translational Medicine | 2017
Ina Chen; Lesley Mathews-Greiner; Dandan Li; Abisola Abisoye-Ogunniyan; Satyajit Ray; Yansong Bian; Vivek Shukla; Xiaohu Zhang; Raj Guha; Craig J. Thomas; Berkley Gryder; Athina Zacharia; Joal D. Beane; Sarangan Ravichandran; Marc Ferrer; Udo Rudloff
BackgroundPatients with hereditary diffuse gastric cancer (HDGC), a cancer predisposition syndrome associated with germline mutations of the CDH1 (E-cadherin) gene, have few effective treatment options. Despite marked differences in natural history, histopathology, and genetic profile to patients afflicted by sporadic gastric cancer, patients with HDGC receive, in large, identical systemic regimens. The lack of a robust preclinical in vitro system suitable for effective drug screening has been one of the obstacles to date which has hampered therapeutic advances in this rare disease.MethodsIn order to identify therapeutic leads selective for the HDGC subtype of gastric cancer, we compared gene expression profiles and drug phenotype derived from an oncology library of 1912 compounds between gastric cancer cells established from a patient with metastatic HDGC harboring a c.1380delA CDH1 germline variant and sporadic gastric cancer cells.ResultsUnsupervised hierarchical cluster analysis shows select gene expression alterations in c.1380delA CDH1 SB.mhdgc-1 cells compared to a panel of sporadic gastric cancer cell lines with enrichment of ERK1–ERK2 (extracellular signal regulated kinase) and IP3 (inositol trisphosphate)/DAG (diacylglycerol) signaling as the top networks in c.1380delA SB.mhdgc-1 cells. Intracellular phosphatidylinositol intermediaries were increased upon direct measure in c.1380delA CDH1 SB.mhdgc-1 cells. Differential high-throughput drug screening of c.1380delA CDH1 SB.mhdgc-1 versus sporadic gastric cancer cells identified several compound classes with enriched activity in c.1380 CDH1 SB.mhdgc-1 cells including mTOR (Mammalian Target Of Rapamycin), MEK (Mitogen-Activated Protein Kinase), c-Src kinase, FAK (Focal Adhesion Kinase), PKC (Protein Kinase C), or TOPO2 (Topoisomerase II) inhibitors. Upon additional drug response testing, dual PI3K (Phosphatidylinositol 3-Kinase)/mTOR and topoisomerase 2A inhibitors displayed up to >100-fold increased activity in hereditary c.1380delA CDH1 gastric cancer cells inducing apoptosis most effectively in cells with deficient CDH1 function.ConclusionIntegrated pharmacological and transcriptomic profiling of hereditary diffuse gastric cancer cells with a loss-of-function c.1380delA CDH1 mutation implies various pharmacological vulnerabilities selective to CDH1-deficient familial gastric cancer cells and suggests novel treatment leads for future preclinical and clinical treatment studies of familial gastric cancer.
Science Translational Medicine | 2018
Marielle E. Yohe; Berkley Gryder; Jack F. Shern; Young K. Song; Hsien-Chao Chou; Sivasish Sindiri; Arnulfo Mendoza; Rajesh Patidar; Xiaohu Zhang; Rajarashi Guha; Donna Butcher; Kristine A. Isanogle; Christina Robinson; Xiaoling Luo; Jin-Qiu Chen; Ashley Walton; Parirokh Awasthi; Elijah F. Edmondson; Simone Difilippantonio; Jun S. Wei; Keji Zhao; Marc Ferrer; Craig J. Thomas; Javed Khan
MEK inhibition decreases cell viability and induces differentiation in RAS-driven rhabdomyosarcoma via an epigenetic mechanism. Muscling in on RAS Rhabdomyosarcomas occur when skeletal muscle precursors fail to differentiate. These tumors often display mutated RAS, although the mechanism through which RAS promotes tumor formation has remained unclear. Yohe et al. discovered that RAS blocks myogenesis in fusion-negative rhabdomyosarcoma (FN-RMS) by inducing epigenetic changes via the MAP kinase pathway. Treatment with the MEK/ERK inhibitor trametinib led to chromatin and super-enhancer remodeling, which in turn resulted in cellular differentiation in cell lines and improved tumor volume and survival in xenograft mouse models. Combining trametinib and an IGF1R inhibitor synergistically improved treatment efficacy in the xenografted mice. The RAS isoforms are frequently mutated in many types of human cancers, including PAX3/PAX7 fusion-negative rhabdomyosarcoma. Pediatric RMS arises from skeletal muscle progenitor cells that have failed to differentiate normally. The role of mutant RAS in this differentiation blockade is incompletely understood. We demonstrate that oncogenic RAS, acting through the RAF–MEK [mitogen-activated protein kinase (MAPK) kinase]–ERK (extracellular signal–regulated kinase) MAPK effector pathway, inhibits myogenic differentiation in rhabdomyosarcoma by repressing the expression of the prodifferentiation myogenic transcription factor, MYOG. This repression is mediated by ERK2-dependent promoter-proximal stalling of RNA polymerase II at the MYOG locus. Small-molecule screening with a library of mechanistically defined inhibitors showed that RAS-driven RMS is vulnerable to MEK inhibition. MEK inhibition with trametinib leads to the loss of ERK2 at the MYOG promoter and releases the transcriptional stalling of MYOG expression. MYOG subsequently opens chromatin and establishes super-enhancers at genes required for late myogenic differentiation. Furthermore, trametinib, in combination with an inhibitor of IGF1R, potently decreases rhabdomyosarcoma cell viability and slows tumor growth in xenograft models. Therefore, this combination represents a potential therapeutic for RAS-mutated rhabdomyosarcoma.
Oncogene | 2018
Timothy McKinnon; Rosemarie Venier; Marielle E. Yohe; Sivasish Sindiri; Berkley Gryder; Jack F. Shern; Leah Kabaroff; Brendan C. Dickson; Krista Schleicher; Guillaume Chouinard-Pelletier; Serena Menezes; Abha A. Gupta; Xiaohu Zhang; Rajarashi Guha; Marc Ferrer; Craig J. Thomas; Yuhong Wei; Dariush Davani; Cynthia J. Guidos; Javed Khan; Rebecca Gladdy
Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma and outcomes have stagnated, highlighting a need for novel therapies. Genomic analysis of RMS has revealed that alterations in the receptor tyrosine kinase (RTK)/RAS/PI3K axis are common and that FGFR4 is frequently mutated or overexpressed. Although FGFR4 is a potentially druggable receptor tyrosine kinase, its functions in RMS are undefined. This study tested FGFR4-activating mutations and overexpression for the ability to generate RMS in mice. Murine tumor models were subsequently used to discover potential therapeutic targets and to test a dual PI3K/mTOR inhibitor in a preclinical setting. Specifically, we provide the first mechanistic evidence of differential potency in the most common human RMS mutations, V550E or N535K, compared to FGFR4wt overexpression as murine myoblasts expressing FGFR4V550E undergo higher rates of cellular transformation, engraftment into mice, and rapidly form sarcomas that highly resemble human RMS. Murine tumor cells overexpressing FGFR4V550E were tested in an in vitro dose–response drug screen along with human RMS cell lines. Compounds were grouped by target class, and potency was determined using average percentage of area under the dose–response curve (AUC). RMS cells were highly sensitive to PI3K/mTOR inhibitors, in particular, GSK2126458 (omipalisib) was a potent inhibitor of FGFR4V550E tumor-derived cell and human RMS cell viability. FGFR4V550E-overexpressing myoblasts and tumor cells had low nanomolar GSK2126458 EC50 values. Mass cytometry using mouse and human RMS cell lines validated GSK2126458 specificity at single-cell resolution, decreasing the abundance of phosphorylated Akt as well as decreasing phosphorylation of the downstream mTOR effectors 4ebp1, Eif4e, and S6. Moreover, PI3K/mTOR inhibition also robustly decreased the growth of RMS tumors in vivo. Thus, by developing a preclinical platform for testing novel therapies, we identified PI3K/mTOR inhibition as a promising new therapy for this devastating pediatric cancer.