Melinda Broward
University of Kansas
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Melinda Broward.
Molecular Cancer Therapeutics | 2013
Ziyan Y. Pessetto; Scott Weir; Geetika Sethi; Melinda Broward; Andrew K. Godwin
Despite significant treatment advances over the past decade, metastatic gastrointestinal stromal tumor (GIST) remains largely incurable. Rare diseases, such as GIST, individually affect small groups of patients but collectively are estimated to affect 25 to 30 million people in the United States alone. Given the costs associated with the discovery, development, and registration of new drugs, orphan diseases such as GIST are often not pursued by mainstream pharmaceutical companies. As a result, “drug repurposing” or “repositioning,” has emerged as an alternative to the traditional drug development process. In this study, we screened 796 U.S. Food and Drug Administration (FDA)-approved drugs and found that two of these compounds, auranofin (Ridaura) and fludarabine phosphate, effectively and selectively inhibited the proliferation of GISTs, including imatinib-resistant cells. One of the most notable drug hits, auranofin, an oral, gold-containing agent approved by the FDA in 1985 for the treatment of rheumatoid arthritis, was found to inhibit thioredoxin reductase activity and induce reactive oxygen species (ROS) production, leading to dramatic inhibition of GIST cell growth and viability. Importantly, the anticancer activity associated with auranofin was independent of imatinib-resistant status, but was closely related to the endogenous and inducible levels of ROS. Coupled with the fact that auranofin has an established safety profile in patients, these findings suggest for the first time that auranofin may have clinical benefit for patients with GIST, particularly in those suffering from imatinib-resistant and recurrent forms of this disease. Mol Cancer Ther; 12(7); 1299–309. ©2013 AACR.
Toxicology and Applied Pharmacology | 2012
Bo Kong; Iván L. Csanaky; Lauren M. Aleksunes; Meghan Patni; Qi Chen; Xiaochao Ma; Hartmut Jaeschke; Scott Weir; Melinda Broward; Curtis D. Klaassen; Grace L. Guo
Emerging evidence suggests that feeding a high-fat diet (HFD) to rodents affects the expression of genes involved in drug transport. However, gender-specific effects of HFD on drug transport are not known. The multidrug resistance-associated protein 2 (Mrp2, Abcc2) is a transporter highly expressed in the hepatocyte canalicular membrane and is important for biliary excretion of glutathione-conjugated chemicals. The current study showed that hepatic Mrp2 expression was reduced by HFD feeding only in female, but not male, C57BL/6J mice. In order to determine whether down-regulation of Mrp2 in female mice altered chemical disposition and toxicity, the biliary excretion and hepatotoxicity of the Mrp2 substrate, α-naphthylisothiocyanate (ANIT), were assessed in male and female mice fed control diet or HFD for 4weeks. ANIT-induced biliary injury is a commonly used model of experimental cholestasis and has been shown to be dependent upon Mrp2-mediated efflux of an ANIT glutathione conjugate that selectively injures biliary epithelial cells. Interestingly, HFD feeding significantly reduced early-phase biliary ANIT excretion in female mice and largely protected against ANIT-induced liver injury. In summary, the current study showed that, at least in mice, HFD feeding can differentially regulate Mrp2 expression and function and depending upon the chemical exposure may enhance or reduce susceptibility to toxicity. Taken together, these data provide a novel interaction between diet and gender in regulating hepatobiliary excretion and susceptibility to injury.
PLOS ONE | 2016
Kishore Polireddy; Ruochen Dong; Peter R. McDonald; Tao Wang; Brendan Luke; Ping Chen; Melinda Broward; Anuradha Roy; Qi Chen
Background Pancreatic cancer has an enrichment of stem-like cancer cells (CSCs) that contribute to chemoresistant tumors prone to metastasis and recurrence. Drug screening assays based on cytotoxicity cannot identify specific CSC inhibitors, because CSCs comprise only a small portion of cancer cell population, and it is difficult to propagate stable CSC populations in vitro for high-throughput screening (HTS) assays. Based on the important role of cancer cell epithelial-to-mesenchymal transition (EMT) in promoting CSCs, we hypothesized that inhibition of EMT can be a useful strategy for inhibiting CSCs, and therefore a feasible approach for HTS can be built for identification of CSC inhibitors, based on assays detecting EMT inhibition. Methods An immunofluorescent assay was established and optimized for HTS to identify compounds that enhance E-cadherin expression, as a hallmark of inhibition of EMT. Four chemical libraries containing 41,472 compounds were screened in PANC-1 pancreatic cancer cell line. Positive hits were validated for EMT and CSC inhibition in vitro using sphere formation assay, western blotting, immune fluorescence, and scratch assay. Results Initial hits were refined to 73 compounds with a secondary screening, among which 17 exhibited concentration dependent induction of E-cadherin expression. Six compounds were selected for further study which belonged to 2 different chemical structural clusters. A novel compound 1-(benzylsulfonyl) indoline (BSI, Compound #38) significantly inhibited pancreatic cancer cell migration and invasion. BSI inhibited histone deacetylase, increased histone 4 acetylation preferably, resulting in E-cadherin up-regulation. BSI effectively inhibited tumor spheres formation. Six more analogues of BSI were tested for anti-migration and anti-CSC activities. Conclusion This study demonstrated a feasible approach for discovery of agents targeting EMT and CSCs using HTS, and identified a class of novel chemicals that could be developed as anti-EMT and anti-CSC drug leads.
PLOS ONE | 2018
Alejandro Parrales; Peter R. McDonald; Megan Ottomeyer; Anuradha Roy; Frank J. Shoenen; Melinda Broward; Tyce Bruns; Douglas H. Thamm; Scott Weir; Kathleen Neville; Tomoo Iwakuma; Joy M. Fulbright
Background Osteosarcoma is an orphan disease for which little improvement in survival has been made since the late 1980s. New drug discovery for orphan diseases is limited by the cost and time it takes to develop new drugs. Repurposing already approved FDA-drugs can help overcome this limitation. Another limitation of cancer drug discovery is the lack of preclinical models that accurately recapitulate what occurs in humans. For OS using dogs as a model can minimize this limitation as OS in canines develops spontaneously, is locally invasive and metastasizes to the lungs as it does in humans. Methods In our present work we used high-throughput screens to identify drugs from a library of 2,286 FDA-approved drugs that demonstrated selective growth inhibition against both human and canine OS cell lines. The identified lead compound was then tested for synergy with 7 other drugs that have demonstrated activity against OS. These results were confirmed with in vitro assays and an in vivo murine model of OS. Results We identified 13 drugs that demonstrated selective growth inhibition against both human and canine OS cell lines. Auranofin was selected for further in vitro combination drug screens. Auranofin showed synergistic effects with vorinostat and rapamycin on OS viability and apoptosis induction. Auranofin demonstrated single-agent growth inhibition in both human and canine OS xenografts, and cooperative growth inhibition was observed in combination with rapamycin or vorinostat. There was a significant decrease in Ki67-positive cells and an increase in cleaved caspase-3 levels in tumor tissues treated with a combination of auranofin and vorinostat or rapamycin. Conclusions Auranofin, alone or in combination with rapamycin or vorinostat, may be useful new treatment strategies for OS. Future studies may evaluate the efficacy of auranofin in dogs with OS as a prelude to human clinical evaluation.
Cancer Research | 2015
Joy M. Fulbright; Kathleen Neville; Melinda Broward; Tyce Bruns; Anuradha Roy; Peter R. McDonald; Megan Ottomeyer; Douglas H. Thamm; Tomoo Iwakuma
Introduction: The purpose of this study was to identify FDA approved drugs that initiate cell death or growth arrest in canine and human osteosarcoma (OS) cell lines in vitro followed by confirmation of anticancer activity in these cell lines. The ultimate objective of this comparative oncology project is to identify promising agents for the treatment of osteosarcoma in dogs and humans, a disease in which survival rates have remained low and stagnant for the last three decades. Procedures: A library of 2,328 FDA approved drugs was screened for activity in 2 canine and 2 human OS cell lines, as well as in 1 corresponding normal cell line. Cell viability was measure using the Cell Titer-Glo™ Luminescent Cell Viability Assay (Promega). FDA approved drugs meeting pre-determined screening criteria for activity, selectivity, and potency were selected for confirmatory studies. Effects on cell proliferation were confirmed using cell counting using trypan blue exclusion performed over a 10 day period. Effects on cell cycle and apoptosis were performed using propidium iodide staining and flow cytometry. Summary: Nine FDA approved drugs were identified based on activity and selectivity. Auranofin, a FDA approved rheumatoid arthritis agent, was selected for cell proliferation and cell cycle evaluation due to its IC50 value in control cells in comparison to OS cell lines. Auranofin displayed cytostatic effects in canine and human OS cell lines at low doses and drugs appeared to induce G2 arrest in treated cells. Conclusions: Auranofin is an FDA approved drug that demonstrates promising, selective anticancer activity in canine and human OS cell lines. The next steps are to determine whether this agent possesses additive or synergistic activity in combination with anticancer agents that have already demonstrated clinical activity in dogs and humans with OS. In vivo preclinical proof of concept will be determined in mouse xenograft studies and dogs with spontaneous OS for promising drug combinations. Subsequently, we hope to rapidly translate preclinical proof of concept findings to human OS patients. Citation Format: Joy M. Fulbright, Kathleen Neville, Melinda Broward, Tyce A. Bruns, Anuradha Roy, Peter McDonald, Megan Ottomeyer, Douglas H. Thamm, Tomoo Iwakuma. Comparative oncology drug discovery for Osteosarcoma in dogs and humans. [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 3689. doi:10.1158/1538-7445.AM2015-3689
Journal of the American Chemical Society | 2007
Wei Jin; John D. Trzupek; Thomas J. Rayl; Melinda Broward; George Vielhauer; Scott Weir; Inkyu Hwang; Dale L. Boger
Journal of Medicinal Chemistry | 2010
James P. Lajiness; William M. Robertson; Irene Dunwiddie; Melinda Broward; George Vielhauer; Scott Weir; Dale L. Boger
Annals of Surgical Oncology | 2010
Mark S. Cohen; Mazin Al-Kasspooles; Stephen K. Williamson; David W. Henry; Melinda Broward; Katherine F. Roby
Cancer Research | 2018
Tomoo Iwakuma; Alejandro Parrales; Peter R. McDonald; Anuradha Roy; Mitchell W. Braun; Frank J. Schoenen; Jenna Wang; Steve Rogers; Melinda Broward; Tyce Bruns; Shrikant Anant; Dan A. Dixon; Fred Meyer; Katherine Chastain; Douglas H. Thamm; Scott Weir; Kathleen Neville; Joy M. Fulbright
Neuro-oncology | 2016
Sara Taylor; Corbin Rayfield; Albert H. Kim; Melinda Broward; Anu Roy; Scott Weir; Kristin R. Swanson; Joshua B. Rubin