Network


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

Hotspot


Dive into the research topics where Anjali Singh is active.

Publication


Featured researches published by Anjali Singh.


Neuro-oncology | 2014

A novel C19MC amplified cell line links Lin28/let-7 to mTOR signaling in embryonal tumor with multilayered rosettes

Tara Spence; Christian Perotti; Patrick Sin-Chan; Daniel Picard; Wei Wu; Anjali Singh; Colleen Anderson; Michael D. Blough; J. Gregory Cairncross; Lucie Lafay-Cousin; Douglas Strother; Cynthia Hawkins; Aru Narendran; Annie Huang; Jennifer A. Chan

BACKGROUND Embryonal tumor with multilayered rosettes (ETMR) is an aggressive central nervous system primitive neuroectodermal tumor (CNS-PNET) variant. ETMRs have distinctive histology, amplification of the chromosome 19 microRNA cluster (C19MC) at chr19q13.41-42, expression of the RNA binding protein Lin28, and dismal prognosis. Functional and therapeutic studies of ETMR have been limited by a lack of model systems. METHODS We have established a first cell line, BT183, from a case of ETMR and characterized its molecular and cellular features. LIN28 knockdown was performed in BT183 to examine the potential role of Lin28 in regulating signaling pathway gene expression in ETMR. Cell line findings were corroborated with immunohistochemical studies in ETMR tissues. A drug screen of 73 compounds was performed to identify potential therapeutic targets. RESULTS The BT183 line maintains C19MC amplification, expresses C19MC-encoded microRNAs, and is tumor initiating. ETMRs, including BT183, have high LIN28 expression and low let-7 miRNA expression, and show evidence of mTOR pathway activation. LIN28 knockdown increases let-7 expression and decreases expression of IGF/PI3K/mTOR pathway components. Pharmacologic inhibition of the mTOR pathway reduces BT183 cell viability. CONCLUSIONS BT183 retains key genetic and histologic features of ETMR. In ETMR, Lin28 is not only a diagnostic marker but also a regulator of genes involved in growth and metabolism. Our findings indicate that inhibitors of the IGF/PI3K/mTOR pathway may be promising novel therapies for these fatal embryonal tumors. As the first patient-derived cell line of these rare tumors, BT183 is an important, unique reagent for investigating ETMR biology and therapeutics.


Pediatric Blood & Cancer | 2011

Cytotoxicity, drug combinability, and biological correlates of ABT‐737 against acute lymphoblastic leukemia cells with MLL rearrangement

Aarthi Jayanthan; Andrea Incoronato; Anjali Singh; Christopher Blackmore; Delphine Bernoux; Victor Lewis; Ronald W. Stam; James A. Whitlock; Aru Narendran

ABT‐737 is a BH3 mimetic small‐molecule inhibitor that binds with high affinity to Bcl‐2 to induce apoptosis in malignant cells and has shown promise as an effective anti‐leukemic agent in pediatric preclinical tests. This study focuses on the effects of ABT‐737 on leukemia cells with MLL rearrangement and identifies some of the biological correlates of its activity.


Molecular Oncology | 2013

Profiling pathway-specific novel therapeutics in preclinical assessment for central nervous system atypical teratoid rhabdoid tumors (CNS ATRT): Favorable activity of targeting EGFR- ErbB2 signaling with lapatinib

Anjali Singh; Xueqing Lun; Aarthi Jayanthan; Halah Obaid; Yibing Ruan; Douglas Strother; Susan N. Chi; Amy Smith; Peter A. Forsyth; Aru Narendran

Despite intensifying multimodal treatments, children with central nervous system atypical teratoid/rhabdoid tumor (CNS ATRT) continue to endure unacceptably high mortality rates. At present, concerted efforts are focusing on understanding the characteristic INI1 mutation and its implications for the growth and survival of these tumors. Additionally, pharmaceutical pipeline libraries constitute a significant source of potential agents that can be taken to clinical trials in a timely manner. However, this process requires efficient target validation and relevant preclinical studies. As an initial screening approach, a panel of 129 small molecule inhibitors from multiple pharmaceutical pipeline libraries was tested against three ATRT cell lines by in vitro cytotoxicity assays. Based on these data, agents that have strong activity and corresponding susceptible cellular pathways were identified. Target modulation, antibody array analysis, drug combination and in vivo xenograft studies were performed on one of the pathway inhibitors found in this screening. Approximately 20% of agents in the library showed activity with IC50 values of 1 μM or less and many showed IC50 values less than 0.05 μM. Intra cell line variability was also noted among some of the drugs. However, it was determined that agents capable of affecting pathways constituting ErbB2, mTOR, proteasomes, Hsp90, Polo like kinases and Aurora kinases were universally effective against the three ATRT cell lines. The first target selected for further analysis, the inhibition of ErbB2‐EGFR pathway by the small molecule inhibitor lapatinib, indicated inhibition of cell migration properties and the initiation of apoptosis. Synergy between lapatinib and IGF‐IR inhibition was also demonstrated by combination index (CI) values. Xenograft studies showed effective antitumor activity of lapatinib in vivo. We present an experimental approach to identifying agents and drug combinations for future clinical trials and provide evidence for the potential of lapatinib as an effective agent in the context of the biology and heterogeneity of its targets in ATRT.


Cellular Physiology and Biochemistry | 2011

Curcumin Blocks Kv11.1 (erg) Potassium Current and Slows Proliferation in the Infant Acute Monocytic Leukemia Cell line THP-1

Umberto Banderali; Darrell D. Belke; Anjali Singh; Aarthi Jayanthan; Wayne R. Giles; Aru Narendran

Acute Myeloid Leukemia (AML) accounts for approximately one fifth of all childhood leukemia yet is responsible for a significant proportion of morbidity and mortality in this population. For this reason, research to identify novel targets for the development of effective AML therapeutics has intensified in the recent past. The THP-1 cell line, which was originally established from an infant diagnosed with AML, provides an experimental model for functional, pre-clinical therapeutics and target identification studies of AML. Here we show the expression of the voltage gated potassium channel Kv11.1 in THP-1 cells as opposed to normal hematopoietic stem cells. In addition, curcumin, a natural polyphenol derived from the plant Curcuma longa, effectively blocked Kv11.1 activity and also inhibited the proliferation of these cells. Curcumin was rapidly internalized by THP-1 cells and possibly exerts potential growth inhibitory activity by interacting with intracellular epitopes of the ion channel. Inhibition of ionic currents carried by Kv11.1 resulted in depolarization of cell membrane potential. We propose that the inhibition of Kv11.1 activity by curcumin may lead to interference with leukemic cell physiology and consequently the suppression of survival and proliferation of AML cells.


Molecular Oncology | 2013

Double-deleted vaccinia virus in virotherapy for refractory and metastatic pediatric solid tumors.

Xueqing Lun; Yibing Ruan; Aarthi Jayanthan; David Liu; Anjali Singh; Tanya M. Trippett; John C. Bell; Peter A. Forsyth; Randal N. Johnston; Aru Narendran

Previous studies have shown successful antitumor effects of systemically delivered double‐deleted vaccinia virus (vvDD) against a number of adult tumor models, including glioma, colon and ovarian cancers. The purpose of this study was to investigate the oncolytic potential of vvDD against a panel of cell lines representative of pediatric solid tumors that are currently difficult to cure.


Leukemia Research | 2011

Induction of apoptosis in pediatric acute lymphoblastic leukemia (ALL) cells by the therapeutic opioid methadone and effective synergy with Bcl-2 inhibition.

Anjali Singh; Aarthi Jayanthan; Allyson Farran; Adam N. Elwi; Sung-Woo Kim; Peter Farran; Aru Narendran

Although recent decades have seen a significant improvement in the treatment outcome of leukemia in the pediatric population, those who are treated for relapsed disease still face significant morbidity and mortality. However, current salvage regimens are often assembled with agents that have similar mode of activity as the chemotherapeutics used in the initial treatment. Hence, novel therapeutic agents that are capable of distinct and diverse mechanisms of activity in, now resistant, leukemia cells are of great interest. We have investigated the opioid agonist methadone for its anti-leukemic activity, initially reported in studies with cell lines derived from adult patients. Our findings show that, compared to normal cells, methadone has enhanced cytotoxicity against specimens and cell lines established from refractory childhood leukemia. In addition, methadones activity synergized with that of the anti-Bcl-2 agent ABT-737 and was characterized by the induction of distinct changes in tumor cell mitochondria. Data presented also identify biological correlates and a potential mechanism for methadone activity by its effects on Mcl-1 and other members of the apoptosis cascade. We provide mechanistic data for the therapeutic potential of a family of agents that is largely unexplored for anti-leukemic activity.


Journal of Experimental & Clinical Cancer Research | 2015

Targeted inhibition of MEK1 by cobimetinib leads to differentiation and apoptosis in neuroblastoma cells

Anjali Singh; Yibing Ruan; Tanya Tippett; Aru Narendran

BackgroundNeuroblastoma (NB) is one of the most common childhood malignancies. Currently, high risk NB carries a poor outcome and significant treatment related toxicities and, thus has been a focus for new therapeutics research in pediatric oncology. In this study, we evaluated the effects of the MEK inhibitor cobimetinib, as a single agent and in combinations, on the growth, survival and differentiation properties against a molecularly representative panel of NB cell lines.MethodsIn vitro anti-proliferative activity of cobimetinib alone or in combination was investigated by cell viability assays and its target modulatory activity was evaluated using phospho-kinases antibody arrays and western blot analysis. To determine the effect of combination with cis-RA on differentiation and resulting enhanced cellular cytotoxicity, the expression of glial fibrillary acidic protein (GFAP) and microtubule-associated protein 2 (MAP2) expression levels were examined by immuno-fluorescence.ResultsOur findings show that cobimetinib alone induced a concentration-dependent loss of cell viability in all NB cell lines. In addition, cobimetinib showed feedback activation of MEK1/2, and the dephosphorylation of extracellular signal-regulated kinases (ERK1/2) and c-RAF, providing information on the biological correlates of MEK inhibition in NB. Combined treatment with cis-RA, led to differentiation and enhanced sensitization of NB cells lines to cobimetinib.ConclusionCollectively, our results provide evidence that cobimetinib, in combination with cis-RA, represents a feasible option to develop novel treatment strategies for refractory NB.


Current Cancer Drug Targets | 2017

In Vitro Sensitivity Profiling of Neuroblastoma Cells Against A Comprehensive Small Molecule Kinase Inhibitor Library to Identify Agents for Future Therapeutic Studies

Anjali Singh; Vanessa Meier-Stephenson; Aarthi Jayanthan; Aru Narendran

BACKGROUND Neuroblastoma (NB) constitutes about 8% of all childhood tumors, yet accounts for more than 15% of deaths, with an unacceptable overall survival rate. These rates are despite the current multimodal therapeutic approaches involving surgery, radiation, chemotherapy and myeloablation with hematopoietic stem cell rescue. Hence, efforts have intensified to identify new targets and novel therapeutic approaches to improve cure rates in these children. Numerous new agents for adult malignancies are developed and evaluated for cancer each year, providing an invaluable resource, with the added advantage of available pharmacologic and toxicity data for consideration. METHODS To identify potential therapeutic targets, we screened a small molecule library of 151 small kinase inhibitors against NB cell lines. Based on our initial screening data, we further examined the potential of Bcr-Abl targeting small molecule inhibitors to affect the growth and survival of NB cells. RESULTS There is diverse activity among the currently available Bcr-Abl inhibitors, possibly reflecting the molecular heterogeneity and off-target activity in each combination. In depth analyses of ponatinib, an oral multi-target kinase inhibitor and effective agent in the treatment of refractory Philadelphia chromosome (Ph) positive leukemia, show growth inhibition at sub-micromolar concentrations. In addition, we also identified the potential of this agent to interfere with insulin-like growth factor-1 receptor (IGF-1R) signaling pathways and Src activity, inhibit cell migration and induce apoptosis. CONCLUSION Our findings provide initial data on ponatinibs potential to target key growth regulatory pathways and provide the rationale for further studies and evaluation in future early phase clinical trials for the treatment of refractory NB.


Cellular Physiology and Biochemistry | 2011

Contents Vol. 28, 2011

Vladimír Štrbák; Kathrine Eduardsen; Susanne L. Larsen; Ivana Novak; Ian Henry Lambert; Else K. Hoffmann; Stine F. Pedersen; Ang Li; Juni Banerjee; Chi Ting Leung; Kim Peterson-Yantorno; W. Daniel Stamer; Mortimer M. Civan; Dorette Z. Ellis; Andreas Schedlbauer; Rosaria Gandini; Georg Kontaxis; Markus Paulmichl; Johannes Fürst; Robert Konrat; Kenneth B. Gagnon; Kerri Rios; Eric Delpire; Bonnie L. Blazer-Yost; Brenda J. Blacklock; Stephanie Flaig; Robert L. Bacallao; Boris Martinac; Vincent H. Gattone; Doris Streif


Cellular Physiology and Biochemistry | 2011

THE NA (+)/H+ EXCHANGER NHE-1, A CRUCIAL CELLULAR INTEGRATOR CONTROLLED BY ITS MEMBRANE ENVIRONMENT

Vladimír Štrbák; Kathrine Eduardsen; Susanne L. Larsen; Ivana Novak; Ian Henry Lambert; Else K. Hoffmann; Stine F. Pedersen; Ang Li; Juni Banerjee; Chi Ting Leung; Kim Peterson-Yantorno; W. Daniel Stamer; Mortimer M. Civan; Dorette Z. Ellis; Andreas Schedlbauer; Rosaria Gandini; Georg Kontaxis; Markus Paulmichl; Johannes Fürst; Robert Konrat; Kenneth B. Gagnon; Kerri Rios; Eric Delpire; Bonnie L. Blazer-Yost; Brenda J. Blacklock; Stephanie Flaig; Robert L. Bacallao; Boris Martinac; Vincent H. Gattone; Doris Streif

Collaboration


Dive into the Anjali Singh's collaboration.

Top Co-Authors

Avatar

Aru Narendran

Alberta Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Aarthi Jayanthan

Alberta Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas Strother

Alberta Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Victor Lewis

Alberta Children'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

Juni Banerjee

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge