Jodi A. Muscal
Baylor College of Medicine
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jodi A. Muscal.
Pediatric Blood & Cancer | 2013
Jodi A. Muscal; Patrick A. Thompson; Terzah M. Horton; Ashish M. Ingle; Charlotte H. Ahern; Renee M. McGovern; Joel M. Reid; Igor Espinoza-Delgado; Brenda Weigel; Susan M. Blaney
A pediatric Phase I trial was performed to determine the maximum‐tolerated dose, dose‐limiting toxicities (DLTs), and pharmacokinetics (PK) of vorinostat and bortezomib, in patients with solid tumors.
Clinical Cancer Research | 2013
Patrick A. Thompson; Jodi A. Muscal; Eshini Panditharatna; Maryam Fouladi; Ashish M. Ingle; Charlotte H. Ahern; Joel M. Reid; Tong Lin; Brenda Weigel; Susan M. Blaney
Purpose: Imetelstat is a covalently-lipidated 13-mer thiophosphoramidate oligonucleotide that acts as a potent specific inhibitor of telomerase. It binds with high affinity to the template region of the RNA component of human telomerase (hTERC) and is a competitive inhibitor of telomerase enzymatic activity. The purpose of this study was to determine the recommended phase II dose of imetelstat in children with recurrent or refractory solid tumors. Experimental Design: Imetelstat was administered intravenously more than two hours on days 1 and 8, every 21 days. Dose levels of 225, 285, and 360 mg/m2 were evaluated, using the rolling-six design. Imetelstat pharmacokinetic and correlative biology studies were also performed during the first cycle. Results: Twenty subjects were enrolled (median age, 14 years; range, 3–21). Seventeen were evaluable for toxicity. The most common toxicities were neutropenia, thrombocytopenia, and lymphopenia, with dose-limiting myelosuppression in 2 of 6 patients at 360 mg/m2. Pharmacokinetics is dose dependent with a lower clearance at the highest dose level. Telomerase inhibition was observed in peripheral blood mononuclear cells at 285 and 360 mg/m2. Two confirmed partial responses, osteosarcoma (n = 1) and Ewing sarcoma (n = 1), were observed. Conclusions: The recommended phase II dose of imetelstat given on days 1 and 8 of a 21-day cycle is 285 mg/m2. Clin Cancer Res; 19(23); 6578–84. ©2013 AACR.
Scientific Reports | 2016
Yongfeng Wang; Long Wang; Shan Guan; Wenming Cao; Hao Wang; Zhenghu Chen; Yanling Zhao; Yang Yu; Huiyuan Zhang; Jonathan C. Pang; Sophia L. Huang; Yo Akiyama; Yifan Yang; Wenjing Sun; Xin Xu; Yan Shi; Hong Zhang; Eugene S. Kim; Jodi A. Muscal; Fengmin Lu; Jianhua Yang
ALK receptor tyrosine kinase has been shown to be a therapeutic target in neuroblastoma. Germline ALK activating mutations are responsible for the majority of hereditary neuroblastoma and somatic ALK activating mutations are also frequently observed in sporadic cases of advanced NB. Crizotinib, a first-line therapy in the treatment of advanced non-small cell lung cancer (NSCLC) harboring ALK rearrangements, demonstrates striking efficacy against ALK-rearranged NB. However, crizotinib fails to effectively inhibit the activity of ALK when activating mutations are present within its kinase domain, as with the F1174L mutation. Here we show that a new ALK inhibitor AZD3463 effectively suppressed the proliferation of NB cell lines with wild type ALK (WT) as well as ALK activating mutations (F1174L and D1091N) by blocking the ALK-mediated PI3K/AKT/mTOR pathway and ultimately induced apoptosis and autophagy. In addition, AZD3463 enhanced the cytotoxic effects of doxorubicin on NB cells. AZD3463 also exhibited significant therapeutic efficacy on the growth of the NB tumors with WT and F1174L activating mutation ALK in orthotopic xenograft mouse models. These results indicate that AZD3463 is a promising therapeutic agent in the treatment of NB.
Pediatric Blood & Cancer | 2016
Piti Techavichit; M. John Hicks; Dolores Lopez-Terrada; Norma M. Quintanilla; R. Paul Guillerman; Stephen F. Sarabia; Hadi Sayeed; Jed G. Nuchtern; Arnold C. Paulino; Jodi A. Muscal; M. Fatih Okcu; Murali Chintagumpala
To determine the clinicopathologic and molecular features and outcome of children with mucoepidermoid carcinoma (MEC).
Pediatric Blood & Cancer | 2016
Piti Techavichit; M. John Hicks; Dolores Lopez-Terrada; Norma M. Quintanilla; R. Paul Guillerman; Stephen F. Sarabia; Hadi Sayeed; Jed G. Nuchtern; Arnold C. Paulino; Jodi A. Muscal; M. Fatih Okcu; Murali Chintagumpala
To determine the clinicopathologic and molecular features and outcome of children with mucoepidermoid carcinoma (MEC).
International Journal of Radiation Oncology Biology Physics | 2009
Jodi A. Muscal; Jeremy Y. Jones; Arnold C. Paulino; Alison A. Bertuch; Jack Su; Shiao Y. Woo; Donald H. Mahoney; Murali Chintagumpala
PURPOSE Acute and late changes in magnetic resonance imaging of the pediatric brain have been described after radiotherapy (RT). We report the post-RT neuroimaging changes in the posterior fossa after intensity-modulated RT (IMRT) in children with medulloblastoma and contrast them with those of leptomeningeal disease. METHODS AND MATERIALS We performed a retrospective review of 53 consecutive children with medulloblastoma who were treated with craniospinal RT followed by IMRT to the posterior fossa and chemotherapy between 1997 and 2006. RESULTS After IMRT to the posterior fossa, 8 (15%) of 53 patients developed increased fluid-attenuated inversion-recovery signal changes in the brainstem or cerebellum and patchy, multifocal, nodular contrast enhancement at a median of 6 months. The enhancement superficially resembled leptomeningeal disease. However, the enhancement resolved without intervention at a median of 6 months later. The accompanying fluid-attenuated inversion-recovery signal changes occasionally preceded the enhancement, were often parenchymal in location, and resolved or persisted to a lesser degree. All 8 patients with transient magnetic resonance imaging changes in the posterior fossa were alive at last follow-up. In contrast, leptomeningeal disease occurred in 8 (15%) of our 53 patients at a median of 19.5 months after IMRT completion. Of these 8 patients, 7 demonstrated initial nodular enhancement outside the conformal field, and 7 patients died. CONCLUSION Magnetic resonance imaging changes can occur in the posterior fossa of children treated with IMRT for medulloblastoma. In our experience, these transient changes occur at a characteristic time and location after RT, allowing them to be distinguished from leptomeningeal disease.
Pediatric Blood & Cancer | 2014
Robin E. Norris; Suzanne Shusterman; Lia Gore; Jodi A. Muscal; Margaret E. Macy; Elizabeth Fox; Noah C. Berkowitz; Aby Buchbinder; Rochelle Bagatell
EZN‐2208 is a water‐soluble PEGylated conjugate of the topoisomerase inhibitor SN38, the active metabolite of irinotecan. Compared to irinotecan, EZN‐2208 has a prolonged half‐life permitting extended exposure to SN38. EZN‐2208 has demonstrated clinical tolerability and antitumor activity in adults with advanced solid tumors. This Phase 1 study evaluated the safety, pharmacokinetics, and preliminary antitumor activity of EZN‐2208 in children with relapsed or refractory solid tumors.
Cancer Letters | 2017
Jiaxiong Lu; Shan Guan; Yanling Zhao; Yang Yu; Sarah E. Woodfield; Huiyuan Zhang; Kristine L. Yang; Shayahati Bieerkehazhi; Lin Qi; Xiao-Nan Li; Jerry Gu; Xin Xu; Jingling Jin; Jodi A. Muscal; Tianshu Yang; Guo-Tong Xu; Jianhua Yang
Activating germline mutations of anaplastic lymphoma kinase (ALK) occur in most cases of hereditary neuroblastoma (NB) and the constitutively active kinase activity of ALK promotes cell proliferation and survival in NB. Therefore, ALK kinase is a potential therapeutic target for NB. In this study, we show that the novel ALK inhibitor alectinib effectively suppressed cell proliferation and induces apoptosis in NB cell lines with either wild-type ALK or mutated ALK (F1174L and D1091N) by blocking ALK-mediated PI3K/Akt/mTOR signaling. In addition, alectinib enhanced doxorubicin-induced cytotoxicity and apoptosis in NB cells. Furthermore, alectinib induced apoptosis in an orthotopic xenograft NB mouse model. Also, in the TH-MYCN transgenic mouse model, alectinib resulted in decreased tumor growth and prolonged survival time. These results indicate that alectinib may be a promising therapeutic agent for the treatment of NB.
Oncotarget | 2017
Shayahati Bieerkehazhi; Zhenghu Chen; Yanling Zhao; Yang Yu; Huiyuan Zhang; Sanjeev A. Vasudevan; Sarah E. Woodfield; Ling Tao; Joanna S. Yi; Jodi A. Muscal; Jonathan C. Pang; Shan Guan; Hong Zhang; Jed G. Nuchtern; Hui Li; Huiwu Li; Jianhua Yang
Neuroblastoma (NB) is the most common extracranial solid tumor in children. Aberrant activation of the non-receptor tyrosine kinases Src and c-Abl contributes to the progression of NB. Thus, targeting these kinases could be a promising strategy for NB therapy. In this paper, we report that the potent dual Src/Abl inhibitor bosutinib exerts anti-tumor effects on NB. Bosutinib inhibited NB cell proliferation in a dose-dependent manner and suppressed colony formation ability of NB cells. Mechanistically, bosutinib effectively decreased the activity of Src/Abl and PI3K/AKT/mTOR, MAPK/ERK, and JAK/STAT3 signaling pathways. In addition, bosutinib enhanced doxorubicin (Dox)- and etoposide (VP-16)-induced cytotoxicity in NB cells. Furthermore, bosutinib demonstrated anti-tumor efficacy in an orthotopic xenograft NB mouse model in a similar mechanism as of that in vitro. In summary, our results reveal that Src and c-Abl are potential therapeutic targets in NB and that the novel Src/Abl inhibitor bosutinib alone or in combination with other chemotherapeutic agents may be a valuable therapeutic option for NB patients.
Clinical Cancer Research | 2018
Mari Kogiso; Lin Qi; Frank Braun; Sarah Injac; Linna Zhang; Yuchen Du; Huiyuan Zhang; Frank Y. Lin; Sibo Zhao; Holly Lindsay; Jack Mf Su; Patricia Baxter; Adekunle M. Adesina; Debra Liao; Mark G. Qian; Stacey L. Berg; Jodi A. Muscal; Xiao-Nan Li
Purpose: Pediatric glioblastoma multiforme (pGBM) is a highly aggressive tumor in need of novel therapies. Our objective was to demonstrate the therapeutic efficacy of MLN8237 (alisertib), an orally available selective inhibitor of Aurora A kinase (AURKA), and to evaluate which in vitro model system (monolayer or neurosphere) can predict therapeutic efficacy in vivo. Experimental Design: AURKA mRNA expressions were screened with qRT-PCR. In vitro antitumor effects were examined in three matching pairs of monolayer and neurosphere lines established from patient-derived orthotopic xenograft (PDOX) models of the untreated (IC-4687GBM), recurrent (IC-3752GBM), and terminal (IC-R0315GBM) tumors, and in vivo therapeutic efficacy through log rank analysis of survival times in two models (IC-4687GBM and IC-R0315GBM) following MLN8237 treatment (30 mg/kg/day, orally, 12 days). Drug concentrations in vivo and mechanism of action and resistance were also investigated. Results: AURKA mRNA overexpression was detected in 14 pGBM tumors, 10 PDOX models, and 6 cultured pGBM lines as compared with 11 low-grade gliomas and normal brains. MLN8237 penetrated into pGBM xenografts in mouse brains. Significant extension of survival times were achieved in IC-4687GBM of which both neurosphere and monolayer were inhibited in vitro, but not in IC-R0315GBM of which only neurosphere cells responded (similar to IC-3752GBM). Apoptosis-mediated MLN8237 induced cell death, and the presence of AURKA-negative and CD133+ cells appears to have contributed to in vivo therapy resistance. Conclusions: MLN8237 successfully targeted AURKA in a subset of pGBMs. Our data suggest that combination therapy should aim at AURKA-negative and/or CD133+ pGBM cells to prevent tumor recurrence. Clin Cancer Res; 24(9); 2159–70. ©2018 AACR.