Network


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

Hotspot


Dive into the research topics where Avadhut D. Joshi is active.

Publication


Featured researches published by Avadhut D. Joshi.


Cancer Research | 2010

Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1

Meghan Joyce Seltzer; Bryson D. Bennett; Avadhut D. Joshi; Ping Gao; Ajit G. Thomas; Dana Ferraris; Takashi Tsukamoto; Camilo Rojas; Barbara S. Slusher; Joshua D. Rabinowitz; Chi V. Dang; Gregory J. Riggins

Mutation at the R132 residue of isocitrate dehydrogenase 1 (IDH1), frequently found in gliomas and acute myelogenous leukemia, creates a neoenzyme that produces 2-hydroxyglutarate (2-HG) from α-ketoglutarate (α-KG). We sought to therapeutically exploit this neoreaction in mutant IDH1 cells that require α-KG derived from glutamine. Glutamine is converted to glutamate by glutaminase and further metabolized to α-KG. Therefore, we inhibited glutaminase with siRNA or the small molecule inhibitor bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) and found slowed growth of glioblastoma cells expressing mutant IDH1 compared with those expressing wild-type IDH1. Growth suppression of mutant IDH1 cells by BPTES was rescued by adding exogenous α-KG. BPTES inhibited glutaminase activity, lowered glutamate and α-KG levels, and increased glycolytic intermediates while leaving total 2-HG levels unaffected. The ability to selectively slow growth in cells with IDH1 mutations by inhibiting glutaminase suggests a unique reprogramming of intermediary metabolism and a potential therapeutic strategy.


Journal of Neuro-oncology | 2010

A survey of glioblastoma genomic amplifications and deletions

Shailaja K. Rao; Jennifer Edwards; Avadhut D. Joshi; I-Mei Siu; Gregory J. Riggins

Glioblastoma Multiforme (GBM) is a malignant brain cancer that develops after accumulating genomic DNA damage that often includes gene amplifications and/or deletions. These copy number changes can be a critical step in brain tumor development. To evaluate glioblastoma genomic copy number changes, we determined the genome-wide copy number alterations in 31 GBMs. Illumina Bead Arrays were used to assay 22 GBMs and Digital Karyotyping was used on 8 GBM cell lines and one primary sample. The common amplifications we observed for all 31 samples was GLI/CDK4 (22.6%), MDM2 (12.9%) and PIK3C2B/MDM4 (12.9%). In the 22 GBM tumors, EGFR was amplified in 22.7% of surgical biopsies. The most common homozygously deleted region contained CDKN2A/CDKN2B (p15 and p16) occurring in 29% of cases. This data was compiled and compared to published array CGH studies of 456 cases of GBMs. Pooling our Illumina data with published studies yielded these average amplification rates: EGFR—35.7%, GLI/CDK4—13.4%, MDM2—9.2%, PIK3C2B/MDM4—7.7%, and PDGFRA—7.7%. The CDKN2A/CDKN2B locus was deleted in 46.4% of the combined cases. This study provides a larger assessment of amplifications and deletions in glioblastoma patient populations and shows that several different copy number technologies can produce similar results. The main pathways known to be involved in GBM tumor formation such as p53 control, growth signaling, and cell cycle control are all represented by amplifications or deletions of critical pathway genes. This information is potentially important for formulating targeted therapy in glioblastoma and for planning genomic studies.


Journal of Neuro-oncology | 2009

Glioblastoma cell growth is suppressed by disruption of fibroblast growth factor pathway signaling

Watcharin Loilome; Avadhut D. Joshi; Colette M. J. ap Rhys; Sara Piccirillo; Vescovi L. Angelo; Gary L. Gallia; Gregory J. Riggins

The Fibroblast Growth Factor (FGF) signaling pathway is reported to stimulate glioblastoma (GBM) growth. In this work we evaluated the effect of FGF2, FGF receptor (FGFR), and small molecule inhibition on GBM cells grown in traditional media, or cultured directly in stem-cell media. These lines each expressed the FGFR1, FGFR3 and FGFR4 receptors. Addition of FGF2 ligand showed significant growth stimulation in 8 of 10 cell lines. Disruption of FGF signaling by a neutralizing FGF2 monoclonal antibody and FGFR1 suppression by RNA interference both partially inhibited cell proliferation. Growth inhibition was temporally correlated with a reduction in MAPK signaling. A receptor tyrosine kinase inhibitor with known FGFR/VEGFR activity, PD173074, showed reproducible growth inhibition. Possible mechanisms of growth suppression by PD173074 were implicated by reduced phosphorylation of AKT and MAPK, known oncogenic signal transducers. Subsequent reduction in the cyclin D1, cyclin D2 and CDK4 cell cycle regulators was also observed. Our results indicate that FGF signaling pathway inhibition as a monotherapy will slow, but not arrest growth of glioblastoma cells.


PLOS ONE | 2012

Evaluation of Tyrosine Kinase Inhibitor Combinations for Glioblastoma Therapy

Avadhut D. Joshi; Watcharin Loilome; I-Mei Siu; Betty Tyler; Gary L. Gallia; Gregory J. Riggins

Glioblastoma multiforme (GBM) is the most common intracranial cancer but despite recent advances in therapy the overall survival remains about 20 months. Whole genome exon sequencing studies implicate mutations in the receptor tyrosine kinase pathways (RTK) for driving tumor growth in over 80% of GBMs. In spite of various RTKs being mutated or altered in the majority of GBMs, clinical studies have not been able to demonstrate efficacy of molecular targeted therapies using tyrosine kinase inhibitors in GBMs. Activation of multiple downstream signaling pathways has been implicated as a possible means by which inhibition of a single RTK has been ineffective in GBM. In this study, we sought a combination of approved drugs that would inhibit in vitro and in vivo growth of GBM oncospheres. A combination consisting of gefitinib and sunitinib acted synergistically in inhibiting growth of GBM oncospheres in vitro. Sunitinib was the only RTK inhibitor that could induce apoptosis in GBM cells. However, the in vivo efficacy testing of the gefitinib and sunitinib combination in an EGFR amplified/ PTEN wild type GBM xenograft model revealed that gefitinib alone could significantly improve survival in animals whereas sunitinib did not show any survival benefit. Subsequent testing of the same drug combination in a different syngeneic glioma model that lacked EGFR amplification but was more susceptible to sunitinib in vitro demonstrated no survival benefit when treated with gefitinib or sunitinib or the gefitinib and sunitinib combination. Although a modest survival benefit was obtained in one of two animal models with EGFR amplification due to gefitinib alone, the addition of sunitinib, to test our best in vitro combination therapy, did not translate to any additional in vivo benefit. Improved targeted therapies, with drug properties favorable to intracranial tumors, are likely required to form effective drug combinations for GBM.


Molecular Cancer | 2011

Sodium ion channel mutations in glioblastoma patients correlate with shorter survival

Avadhut D. Joshi; D. Williams Parsons; Victor E. Velculescu; Gregory J. Riggins

BackgroundGlioblastoma Multiforme (GBM) is the most common and invasive astrocytic tumor associated with dismal prognosis. Treatment for GBM patients has advanced, but the median survival remains a meager 15 months. In a recent study, 20,000 genes from 21 GBM patients were sequenced that identified frequent mutations in ion channel genes. The goal of this study was to determine whether ion channel mutations have a role in disease progression and whether molecular targeting of ion channels is a promising therapeutic strategy for GBM patients. Therefore, we compared GBM patient survival on the basis of presence or absence of mutations in calcium, potassium and sodium ion transport genes. Cardiac glycosides, known sodium channel inhibitors, were then tested for their ability to inhibit GBM cell proliferation.ResultsNearly 90% of patients showed at least one mutation in ion transport genes. GBM patients with mutations in sodium channels showed a significantly shorter survival compared to patients with no sodium channel mutations, whereas a similar comparison based on mutational status of calcium or potassium ion channel mutations showed no survival differences. Experimentally, targeting GBM cells with cardiac glycosides such as digoxin and ouabain demonstrated preferential cytotoxicity against U-87 and D54 GBM cells compared to non-tumor astrocytes (NTAs).ConclusionsThese pilot studies of GBM patients with sodium channel mutations indicate an association with a more aggressive disease and significantly shorter survival. Moreover, inhibition of GBM cells by ion channel inhibitors such as cardiac glycosides suggest a therapeutic strategy with relatively safe drugs for targeting GBM ion channel mutations. Key Words: glioblastoma multiforme, ion channels, mutations, small molecule inhibitors, cardiac glycosides.


Neuro-oncology | 2015

Local delivery of cancer-cell glycolytic inhibitors in high-grade glioma

Robert T. Wicks; Javad Azadi; Antonella Mangraviti; Irma Zhang; Lee Hwang; Avadhut D. Joshi; Hansen Bow; Marianne Hütt-Cabezas; Kristin L. Martin; Michelle A. Rudek; Ming Zhao; Henry Brem; Betty Tyler

BACKGROUND 3-bromopyruvate (3-BrPA) and dichloroacetate (DCA) are inhibitors of cancer-cell specific aerobic glycolysis. Their application in glioma is limited by 3-BrPAs inability to cross the blood-brain-barrier and DCAs dose-limiting toxicity. The safety and efficacy of intracranial delivery of these compounds were assessed. METHODS Cytotoxicity of 3-BrPA and DCA were analyzed in U87, 9L, and F98 glioma cell lines. 3-BrPA and DCA were incorporated into biodegradable pCPP:SA wafers, and the maximally tolerated dose was determined in F344 rats. Efficacies of the intracranial 3-BrPA wafer and DCA wafer were assessed in a rodent allograft model of high-grade glioma, both as a monotherapy and in combination with temozolomide (TMZ) and radiation therapy (XRT). RESULTS 3-BrPA and DCA were found to have similar IC50 values across the 3 glioma cell lines. 5% 3-BrPA wafer-treated animals had significantly increased survival compared with controls (P = .0027). The median survival of rats with the 50% DCA wafer increased significantly compared with both the oral DCA group (P = .050) and the controls (P = .02). Rats implanted on day 0 with a 5% 3-BrPA wafer in combination with TMZ had significantly increased survival over either therapy alone. No statistical difference in survival was noted when the wafers were added to the combination therapy of TMZ and XRT, but the 5% 3-BrPA wafer given on day 0 in combination with TMZ and XRT resulted in long-term survivorship of 30%. CONCLUSION Intracranial delivery of 3-BrPA and DCA polymer was safe and significantly increased survival in an animal model of glioma, a potential novel therapeutic approach. The combination of intracranial 3-BrPA and TMZ provided a synergistic effect.


Clinical Neurology and Neurosurgery | 2011

Glioblastoma multiforme in the Muir-Torre syndrome.

Zev A. Binder; Michael W. Johnson; Avadhut D. Joshi; Christine L. Hann; Constance A. Griffin; Alessandro Olivi; Gregory J. Riggins; Gary L. Gallia

Muir-Torre syndrome (MTS) is an autosomal dominant subtype of nonpolyposis colorectal carcinoma (HNPCC) characterized by the development of sebaceous gland tumors and visceral malignancies. The most common subtype of MTS is characterized by germline mutations in mismatch repair (MMR) genes leading to microsatellite instability (MSI). Central nervous system tumors have only rarely been associated with MTS. In this report, we describe the development of a glioblastoma multiforme (GBM) in a patient with MTS. Immunohistochemical analysis of the patients colon carcinoma and his GBM both revealed loss of the mismatch repair proteins mutS homolog 2 (MSH2) and mutS homolog 6 (MSH6).


Oncotarget | 2017

Synergistic and targeted therapy with a procaspase-3 activator and temozolomide extends survival in glioma rodent models and is feasible for the treatment of canine malignant glioma patients

Avadhut D. Joshi; Rachel C. Botham; Lisa J. Schlein; Howard S. Roth; Antonella Mangraviti; Alexandra Borodovsky; Betty Tyler; Steve Joslyn; Jayme S. Looper; Michael Podell; Timothy M. Fan; Paul J. Hergenrother; Gregory J. Riggins

PURPOSE Glioblastoma is a deadly brain cancer with a median survival time of ~15 months. Ionizing radiation plus the DNA alkylator temozolomide (TMZ) is the current standard therapy. PAC-1, a procaspase-3 activating small molecule, is blood-brain barrier penetrant and has previously demonstrated ability to synergize with diverse pro-apoptotic chemotherapeutics. We studied if PAC-1 could enhance the activity of TMZ, and whether addition of PAC-1 to standard treatment would be feasible in spontaneous canine malignant gliomas. EXPERIMENTAL DESIGN Using cell lines and online gene expression data, we identified procaspase-3 as a potential molecular target for most glioblastomas. We investigated PAC-1 as a single agent and in combination with TMZ against glioma cells in culture and in orthotopic rodent models of glioma. Three dogs with spontaneous gliomas were treated with an analogous human glioblastoma treatment protocol, with concurrent PAC-1. RESULTS Procaspase-3 is expressed in gliomas, with higher gene expression correlating with increased tumor grade and decreased prognosis. PAC-1 is cytotoxic to glioma cells in culture and active in orthotopic rodent glioma models. PAC-1 added to TMZ treatments in cell culture increases apoptotic death, and the combination significantly increases survival in orthotopic glioma models. Addition of PAC-1 to TMZ and radiation was well-tolerated in 3 out of 3 pet dogs with spontaneous glioma, and partial to complete tumor reductions were observed. CONCLUSIONS Procaspase-3 is a clinically relevant target for treatment of glioblastoma. Synergistic activity of PAC-1/TMZ in rodent models and the demonstration of feasibility of the combined regime in canine patients suggest potential for PAC-1 in the treatment of glioblastoma.Purpose Glioblastoma is a deadly brain cancer with a median survival time of ∼15 months. Ionizing radiation plus the DNA alkylator temozolomide (TMZ) is the current standard therapy. PAC-1, a procaspase-3 activating small molecule, is blood-brain barrier penetrant and has previously demonstrated ability to synergize with diverse pro-apoptotic chemotherapeutics. We studied if PAC-1 could enhance the activity of TMZ, and whether addition of PAC-1 to standard treatment would be feasible in spontaneous canine malignant gliomas. Experimental Design Using cell lines and online gene expression data, we identified procaspase-3 as a potential molecular target for most glioblastomas. We investigated PAC-1 as a single agent and in combination with TMZ against glioma cells in culture and in orthotopic rodent models of glioma. Three dogs with spontaneous gliomas were treated with an analogous human glioblastoma treatment protocol, with concurrent PAC-1. Results Procaspase-3 is expressed in gliomas, with higher gene expression correlating with increased tumor grade and decreased prognosis. PAC-1 is cytotoxic to glioma cells in culture and active in orthotopic rodent glioma models. PAC-1 added to TMZ treatments in cell culture increases apoptotic death, and the combination significantly increases survival in orthotopic glioma models. Addition of PAC-1 to TMZ and radiation was well-tolerated in 3 out of 3 pet dogs with spontaneous glioma, and partial to complete tumor reductions were observed. Conclusions Procaspase-3 is a clinically relevant target for treatment of glioblastoma. Synergistic activity of PAC-1/TMZ in rodent models and the demonstration of feasibility of the combined regime in canine patients suggest potential for PAC-1 in the treatment of glioblastoma.


Cancer Research | 2015

Abstract 3620: An oral procaspase activating drug, PAC-1, shows preclinical promise for glioblastoma therapy

Avadhut D. Joshi; Rachel C. Botham; Howard S. Roth; Timothy M. Fan; Theodore M. Tarasow; Paul J. Hergenrother; Gregory J. Riggins

PAC-1 is an oral administered procaspase-3 activating drug developed to overcome the malignant cell9s barrier to apoptosis. The promising preclinical safety and efficacy of PAC-1 has led to its consideration for a first in human phase 1 clinical trial for recurrent and advanced malignancies. We have found that because PAC-1 is able to reach intracranial brain tumors in effective concentrations, that it is particularly useful for glioblastoma (GBM), an aggressive and invasive malignancy responsible for the most deaths due to brain cancer. In glioblastoma patients a worse prognosis was correlated with high expression of procaspase-3 mRNA, indicating that procaspase-3 activation may be involved in the pathology of the tumor progression. In preclinical testing of PAC-1, it showed significant survival benefit and low systemic toxicity, in orthotopic animal models of glioblastoma. An aqueous suspension of PAC-1 delivered orally to rats with intracranial 9L glioblastoma improved survival compared to untreated controls. Additionally, when PAC-1 was administered as an oral capsule to rats with 9L glioblastoma, median survival increased over three fold. Oral PAC-1 improved survival significantly also in a human xenograft model (p = 0.04) compared to controls. PAC-1 combined effectively with standard of care, and in some animals, either alone or in combination resulted in long term survival. PAC-1 has the ability to penetrate sufficiently within intracranial tumors and its ability to overcome resistance to apoptosis and activate widespread cell death specifically in the malignant cells makes it a promising small molecule for further preclinical and clinical development. Citation Format: Avadhut D. Joshi, Rachel C. Botham, Howard S. Roth, Timothy M. Fan, Theodore M. Tarasow, Paul J. Hergenrother, Gregory J. Riggins. An oral procaspase activating drug, PAC-1, shows preclinical promise for glioblastoma therapy. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3620. doi:10.1158/1538-7445.AM2015-3620


Archive | 2016

MEBENDAZOLE POLYMORPH FOR TREATMENT AND PREVENTION OF TUMORS

Gregory Riggins; Renyuan Bai; Verena Staedtke; Avadhut D. Joshi; Tara Williamson

Collaboration


Dive into the Avadhut D. Joshi's collaboration.

Top Co-Authors

Avatar

Gregory J. Riggins

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Gary L. Gallia

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Betty Tyler

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

I-Mei Siu

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ajit G. Thomas

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge