Network


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

Hotspot


Dive into the research topics where Shilpi Arora is active.

Publication


Featured researches published by Shilpi Arora.


International Journal of Cancer | 2011

MicroRNA-328 is associated with (non-small) cell lung cancer (NSCLC) brain metastasis and mediates NSCLC migration

Shilpi Arora; Aarati R. Ranade; Nhan L. Tran; Sara Nasser; Shravan Sridhar; Ronald L. Korn; Julianna T.D. Ross; Harshil Dhruv; Kristen M. Foss; Zita Sibenaller; Timothy C. Ryken; Michael B. Gotway; Seungchan Kim; Glen J. Weiss

Brain metastasis (BM) can affect ∼ 25% of nonsmall cell lung cancer (NSCLC) patients during their lifetime. Efforts to characterize patients that will develop BM have been disappointing. microRNAs (miRNAs) regulate the expression of target mRNAs. miRNAs play a role in regulating a variety of targets and, consequently, multiple pathways, which make them a powerful tool for early detection of disease, risk assessment, and prognosis. We investigated miRNAs that may serve as biomarkers to differentiate between NSCLC patients with and without BM. miRNA microarray profiling was performed on samples from clinically matched NSCLC from seven patients with BM (BM+) and six without BM (BM−). Using t‐test and further qRT‐PCR validation, eight miRNAs were confirmed to be significantly differentially expressed. Of these, expression of miR‐328 and miR‐330‐3p were able to correctly classify BM+ vs. BM− patients. This classifier was used on a validation cohort (n = 15), and it correctly classified 12/15 patients. Gene expression analysis comparing A549 parental and A549 cells stably transfected to over‐express miR‐328 (A549‐328) identified several significantly differentially expressed genes. PRKCA was one of the genes over‐expressed in A549‐328 cells. Additionally, A549‐328 cells had significantly increased cell migration compared to A549 cells, which was significantly reduced upon PRKCA knockdown. In summary, miR‐328 has a role in conferring migratory potential to NSCLC cells working in part through PRKCA and with further corroboration in additional independent cohorts, these miRNAs may be incorporated into clinical treatment decision making to stratify NSCLC patients at higher risk for developing BM.


Journal of Translational Medicine | 2009

Synthetic lethal RNAi screening identifies sensitizing targets for gemcitabine therapy in pancreatic cancer

David O. Azorsa; Irma M. Gonzales; Gargi D. Basu; Ashish Choudhary; Shilpi Arora; Kristen M. Bisanz; Jeffrey Kiefer; Meredith C. Henderson; Jeffrey M. Trent; Daniel D. Von Hoff; Spyro Mousses

BackgroundPancreatic cancer retains a poor prognosis among the gastrointestinal cancers. It affects 230,000 individuals worldwide, has a very high mortality rate, and remains one of the most challenging malignancies to treat successfully. Treatment with gemcitabine, the most widely used chemotherapeutic against pancreatic cancer, is not curative and resistance may occur. Combinations of gemcitabine with other chemotherapeutic drugs or biological agents have resulted in limited improvement.MethodsIn order to improve gemcitabine response in pancreatic cancer cells, we utilized a synthetic lethal RNAi screen targeting 572 known kinases to identify genes that when silenced would sensitize pancreatic cancer cells to gemcitabine.ResultsResults from the RNAi screens identified several genes that, when silenced, potentiated the growth inhibitory effects of gemcitabine in pancreatic cancer cells. The greatest potentiation was shown by siRNA targeting checkpoint kinase 1 (CHK1). Validation of the screening results was performed in MIA PaCa-2 and BxPC3 pancreatic cancer cells by examining the dose response of gemcitabine treatment in the presence of either CHK1 or CHK2 siRNA. These results showed a three to ten-fold decrease in the EC50 for CHK1 siRNA-treated cells versus control siRNA-treated cells while treatment with CHK2 siRNA resulted in no change compared to controls. CHK1 was further targeted with specific small molecule inhibitors SB 218078 and PD 407824 in combination with gemcitabine. Results showed that treatment of MIA PaCa-2 cells with either of the CHK1 inhibitors SB 218078 or PD 407824 led to sensitization of the pancreatic cancer cells to gemcitabine.ConclusionThese findings demonstrate the effectiveness of synthetic lethal RNAi screening as a tool for identifying sensitizing targets to chemotherapeutic agents. These results also indicate that CHK1 could serve as a putative therapeutic target for sensitizing pancreatic cancer cells to gemcitabine.


Chemistry & Biology | 2014

EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation.

William D. Bradley; Shilpi Arora; Jennifer Busby; Srividya Balasubramanian; Victor S. Gehling; Christopher G. Nasveschuk; Rishi G. Vaswani; Chih-Chi Yuan; Charlie Hatton; Feng Zhao; Kaylyn E. Williamson; Priyadarshini Iyer; Jacqui Mendez; Robert E. Campbell; Nico Cantone; Shivani Garapaty-Rao; James E. Audia; Andrew Simon Cook; Les A. Dakin; Brian K. Albrecht; Jean-Christophe Harmange; Danette L. Daniels; Richard T. Cummings; Barbara M. Bryant; Emmanuel Normant; Patrick Trojer

The histone lysine methyltransferase (MT) Enhancer of Zeste Homolog 2 (EZH2) is considered an oncogenic driver in a subset of germinal center B-cell-like diffuse large B cell lymphoma (GCB-DLBCL) and follicular lymphoma due to the presence of recurrent, monoallelic mutations in the EZH2 catalytic domain. These genomic data suggest that targeting the EZH2 MT activity is a valid therapeutic strategy for the treatment of lymphoma patients with EZH2 mutations. Here we report the identification of highly potent and selective EZH2 small molecule inhibitors, their validation by a cellular thermal shift assay, application across a large cell panel representing various non-Hodgkins lymphoma (NHL) subtypes, and their efficacy in EZH2mutant-containing GCB-DLBCL xenograft models. Surprisingly, our EZH2 inhibitors selectively affect the turnover of trimethylated, but not monomethylated histone H3 lysine 27 at pharmacologically relevant doses. Importantly, we find that these inhibitors are broadly efficacious also in NHL models with wild-type EZH2.


Nature Chemical Biology | 2016

An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells

Maia Vinogradova; Victor S. Gehling; Amy Gustafson; Shilpi Arora; Charles Tindell; Catherine Wilson; Kaylyn E. Williamson; Gulfem D. Guler; Pranoti Gangurde; Wanda Manieri; Jennifer Busby; E. Megan Flynn; Fei Lan; Hyo-Jin Kim; Shobu Odate; Andrea G. Cochran; Yichin Liu; Matthew Wongchenko; Yibin Yang; Tommy K. Cheung; Tobias M. Maile; Ted Lau; Michael Costa; Ganapati V. Hegde; Erica Jackson; Robert M. Pitti; David Arnott; Christopher M. Bailey; Steve Bellon; Richard T. Cummings

The KDM5 family of histone demethylases catalyzes the demethylation of histone H3 on lysine 4 (H3K4) and is required for the survival of drug-tolerant persister cancer cells (DTPs). Here we report the discovery and characterization of the specific KDM5 inhibitor CPI-455. The crystal structure of KDM5A revealed the mechanism of inhibition of CPI-455 as well as the topological arrangements of protein domains that influence substrate binding. CPI-455 mediated KDM5 inhibition, elevated global levels of H3K4 trimethylation (H3K4me3) and decreased the number of DTPs in multiple cancer cell line models treated with standard chemotherapy or targeted agents. These findings show that pretreatment of cancer cells with a KDM5-specific inhibitor results in the ablation of a subpopulation of cancer cells that can serve as the founders for therapeutic relapse.


Blood | 2012

RNAi screening of the kinome with cytarabine in leukemias

Raoul Tibes; James M Bogenberger; Leena Chaudhuri; R. Tanner Hagelstrom; Donald Chow; Megan E. Buechel; Irma M. Gonzales; Tim Demuth; James L. Slack; Ruben A. Mesa; Esteban Braggio; Hongwei H. Yin; Shilpi Arora; David O. Azorsa

To identify rational therapeutic combinations with cytarabine (Ara-C), we developed a high-throughput, small-interference RNA (siRNA) platform for myeloid leukemia cells. Of 572 kinases individually silenced in combination with Ara-C, silencing of 10 (1.7%) and 8 (1.4%) kinases strongly increased Ara-C activity in TF-1 and THP-1 cells, respectively. The strongest molecular concepts emerged around kinases involved in cell-cycle checkpoints and DNA-damage repair. In confirmatory siRNA assays, inhibition of WEE1 resulted in more potent and universal sensitization across myeloid cell lines than siRNA inhibition of PKMYT1, CHEK1, or ATR. Treatment of 8 acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), and chronic myeloid leukemia (CML) cell lines with commercial and the first-in-class clinical WEE1 kinase inhibitor MK1775 confirmed sensitization to Ara-C up to 97-fold. Ex vivo, adding MK1775 substantially reduced viability in 13 of 14 AML, CML, and myelodysplastic syndrome patient samples compared with Ara-C alone. Maximum sensitization occurred at lower to moderate concentrations of both drugs. Induction of apoptosis was increased using a combination of Ara-C and MK1775 compared with using either drug alone. WEE1 is expressed in primary AML, ALL, and CML specimens. Data from this first siRNA-kinome sensitizer screen suggests that inhibiting WEE1 in combination with Ara-C is a rational combination for the treatment of myeloid and lymphoid leukemias.


The Journal of Clinical Endocrinology and Metabolism | 2010

Detailed Molecular Fingerprinting of Four New Anaplastic Thyroid Carcinoma Cell Lines and Their Use for Verification of RhoB as a Molecular Therapeutic Target

Laura A. Marlow; Jaclyn D'Innocenzi; Yilin Zhang; Stephen D. Rohl; Simon J. Cooper; Thomas J. Sebo; Clive S. Grant; Bryan McIver; Jan L. Kasperbauer; J. Trad Wadsworth; John D. Casler; Pamela W. Kennedy; W. Edward Highsmith; Orlo H. Clark; Dragana Milosevic; Brian C. Netzel; Kendall W. Cradic; Shilpi Arora; Christian Beaudry; Stefan K. Grebe; Marc L. Silverberg; David O. Azorsa; Robert C. Smallridge; John A. Copland

CONTEXT Anaplastic thyroid carcinoma (ATC) is a highly aggressive carcinoma in need of therapeutic options. One critical component of drug discovery is the availability of well-characterized cell lines for identification of molecular mechanisms related to tumor biology and drug responsiveness. Up to 42% of human thyroid cancer cell lines are redundant or not of correct tissue origin, and a comprehensive analysis is currently nonexistent. Mechanistically, RhoB has been identified as a novel molecular target for ATC therapy. OBJECTIVE The aim was to develop four ATC cell lines detailing genetic, molecular, and phenotypic characteristics and to test five classes of drugs on the cell lines to determine whether they inhibited cell proliferation in a RhoB-dependent fashion. DESIGN Four cell lines were derived from ATC tumors. Short tandem DNA repeat and mutational status of the originating tumors and cell lines were performed along with molecular and phenotypic characterizations. Compounds were tested for growth inhibition and ability to up-regulate RhoB. RESULTS Cell line authenticity was confirmed by DNA short tandem repeat analysis. Each proved unique regarding expression of thyroid markers, oncogene status, amplified and deleted genes, and proliferative growth rates. FTI-277, GGTI-286, lovastatin, romidepsin, and UCN-01 up-regulated RhoB and inhibited cell proliferation in a dose-responsive fashion with only romidepsin and FTI-277 being RhoB dependent. CONCLUSIONS Molecular descriptions of thyroid lines were matched to the originating tumors, setting a new standard for cell line characterization. Furthermore, suppressed RhoB is implicated as a molecular target for therapy against ATC because five classes of drugs up-regulate RhoB and inhibit growth dose-responsively.


Lung Cancer | 2012

Hedgehog signaling pathway molecules and ALDH1A1 expression in early-stage non-small cell lung cancer

Guy Raz; Kristi Allen; Chris Kingsley; Irene Cherni; Shilpi Arora; Aprill Watanabe; Carlos D. Lorenzo; David K. Edwards; Shravan Sridhar; Galen Hostetter; Glen J. Weiss

INTRODUCTION The Hedgehog Signaling Pathway (HHSP) has been implicated in the development of multiple cancers. HHSP activation may primarily be hedgehog ligand-dependent in non-small cell lung cancer (NSCLC); while a subset may be ligand-independent. In this study NSCLC primary tumors were used to identify correlations between multiple biomarkers thought to be involved in the HHSP and the clinical outcomes of patients with NSCLC. Identification of such correlations could be used to aid in NSCLC treatment and predicting patient prognosis. METHODS A tissue microarray representing 248 clinically annotated stage I-II NSCLC cases was stained using immunohistochemistry (IHC) and scored for HHSP proteins namely, SHH, PTCH1, SMO, GLI1, and GLI2; as well as, ALDH1A1, a putative cancer stem cell marker. Data was analyzed for correlation between IHC staining, EGFR and KRAS mutations, and clinical characteristics including relapse-free survival (RFS) and overall survival (OS). RESULTS In adenocarcinoma, there were significant correlations of IHC expression between SHH and downstream HHSP receptor SMO (p=0.017) and transcription factor GLI1 (p=0.001), while SMO correlated with GLI1 (p=0.007). In squamous cell carcinoma, SHH significantly correlated with GLI2 protein expression (p=0.003). After multiple testing correction, there was no significant correlation between any of the six markers and RFS or OS. CONCLUSIONS Key downstream components of the HHSP show correlation with sonic hedgehog ligand (SHH) expression, suggesting that ligand-dependent signaling is more prevalent in primary NSCLC tumors. Surprisingly, in early-stage NSCLC, there were no significant correlations between HHSP proteins or ALDH1A1 and RFS or OS.


Cancer Research | 2016

Pharmacological Inhibition of the Histone Lysine Demethylase KDM1A Suppresses the Growth of Multiple Acute Myeloid Leukemia Subtypes.

John P. McGrath; Kaylyn Williamson; Srividya Balasubramanian; Shobu Odate; Shilpi Arora; Charlie Hatton; Thomas M. Edwards; Thomas O'Brien; Steven Magnuson; David Stokoe; Danette L. Daniels; Barbara M. Bryant; Patrick Trojer

Lysine-specific demethylase 1 (KDM1A) is a transcriptional coregulator that can function in both the activation and repression of gene expression, depending upon context. KDM1A plays an important role in hematopoiesis and was identified as a dependency factor in leukemia stem cell populations. Therefore, we investigated the consequences of inhibiting KDM1A in a panel of cell lines representing all acute myelogenous leukemia (AML) subtypes using selective, reversible and irreversible KDM1A small-molecule inhibitors. Cell models of AML, CML, and T-ALL were potently affected by KDM1A inhibition, and cells bearing RUNX1-RUNX1T1 (AML1-ETO) translocations were especially among the most sensitive. RNAi-mediated silencing of KDM1A also effectively suppressed growth of RUNX1-RUNX1T1-containing cell lines. Furthermore, pharmacologic inhibition of KDM1A resulted in complete abrogation of tumor growth in an AML xenograft model harboring RUNX1-RUNX1T1 translocations. We unexpectedly found that KDM1A-targeting compounds not only inhibited the catalytic activity of the enzyme, but evicted KDM1A from target genes. Accordingly, compound-mediated KDM1A eviction was associated with elevated levels of local histone H3 lysine 4 dimethylation, and increased target gene expression, which was further accompanied by cellular differentiation and induction of cell death. Finally, our finding that KDM1A inhibitors effectively synergize with multiple conventional as well as candidate anti-AML agents affords a framework for potential future clinical application. Cancer Res; 76(7); 1975-88. ©2016 AACR.


Molecular Cancer Research | 2011

High-throughput RNAi screening identifies a role for TNK1 in growth and survival of pancreatic cancer cells

Meredith C. Henderson; Irma M. Gonzales; Shilpi Arora; Ashish Choudhary; Jeffrey M. Trent; Daniel D. Von Hoff; Spyro Mousses; David O. Azorsa

To identify novel targets in pancreatic cancer cells, we used high-throughput RNAi (HT-RNAi) to select genes that, when silenced, would decrease viability of pancreatic cancer cells. The HT-RNAi screen involved reverse transfecting the pancreatic cancer cell line BxPC3 with a siRNA library targeting 572 kinases. From replicate screens, approximately 32 kinases were designated as hits, of which 22 kinase targets were selected for confirmation and validation. One kinase identified as a hit from this screen was tyrosine kinase nonreceptor 1 (TNK1), a kinase previously identified as having tumor suppressor-like properties in embryonic stem cells. Silencing of TNK1 with siRNA showed reduced proliferation in a panel of pancreatic cancer cell lines. Furthermore, we showed that silencing of TNK1 led to increased apoptosis through a caspase-dependent pathway and that targeting TNK1 with siRNA can synergize with gemcitabine treatment. Despite previous reports that TNK1 affects Ras and NF-κB signaling, we did not find similar correlations with these pathways in pancreatic cancer cells. Our results suggest that TNK1 in pancreatic cancer cells does not possess the same tumor suppressor properties seen in embryonic cells but seems to be involved in growth and survival. The application of functional genomics by using HT-RNAi screens has allowed us to identify TNK1 as a growth-associated kinase in pancreatic cancer cells. Mol Cancer Res; 9(6); 724–32. ©2011 AACR.


Molecular Cancer | 2010

RNAi phenotype profiling of kinases identifies potential therapeutic targets in Ewing's sarcoma

Shilpi Arora; Irma M. Gonzales; R. Tanner Hagelstrom; Christian Beaudry; Ashish Choudhary; Chao Sima; Raoul Tibes; Spyro Mousses; David O. Azorsa

BackgroundEwings sarcomas are aggressive musculoskeletal tumors occurring most frequently in the long and flat bones as a solitary lesion mostly during the teen-age years of life. With current treatments, significant number of patients relapse and survival is poor for those with metastatic disease. As part of novel target discovery in Ewings sarcoma, we applied RNAi mediated phenotypic profiling to identify kinase targets involved in growth and survival of Ewings sarcoma cells.ResultsFour Ewings sarcoma cell lines TC-32, TC-71, SK-ES-1 and RD-ES were tested in high throughput-RNAi screens using a siRNA library targeting 572 kinases. Knockdown of 25 siRNAs reduced the growth of all four Ewings sarcoma cell lines in replicate screens. Of these, 16 siRNA were specific and reduced proliferation of Ewings sarcoma cells as compared to normal fibroblasts. Secondary validation and preliminary mechanistic studies highlighted the kinases STK10 and TNK2 as having important roles in growth and survival of Ewings sarcoma cells. Furthermore, knockdown of STK10 and TNK2 by siRNA showed increased apoptosis.ConclusionIn summary, RNAi-based phenotypic profiling proved to be a powerful gene target discovery strategy, leading to successful identification and validation of STK10 and TNK2 as two novel potential therapeutic targets for Ewings sarcoma.

Collaboration


Dive into the Shilpi Arora's collaboration.

Top Co-Authors

Avatar

David O. Azorsa

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Irma M. Gonzales

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Patrick Trojer

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar

Meredith C. Henderson

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Spyro Mousses

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ashish Choudhary

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Christian Beaudry

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Glen J. Weiss

Cancer Treatment Centers of America

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John A. Copland

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge