Network


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

Hotspot


Dive into the research topics where Brian A. Menegaz is active.

Publication


Featured researches published by Brian A. Menegaz.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Modeling Ewing sarcoma tumors in vitro with 3D scaffolds

Eliza Li Shan Fong; Salah Eddine Lamhamedi-Cherradi; Emily Burdett; Vandhana Ramamoorthy; Alexander J. Lazar; F. Kurtis Kasper; Mary C. Farach-Carson; Deeksha Vishwamitra; Elizabeth G. Demicco; Brian A. Menegaz; Hesham M. Amin; Antonios G. Mikos; Joseph A. Ludwig

The pronounced biological influence of the tumor microenvironment on cancer progression and metastasis has gained increased recognition over the past decade, yet most preclinical antineoplastic drug testing is still reliant on conventional 2D cell culture systems. Although monolayer cultures recapitulate some of the phenotypic traits observed clinically, they are limited in their ability to model the full range of microenvironmental cues, such as ones elicited by 3D cell–cell and cell–extracellular matrix interactions. To address these shortcomings, we established an ex vivo 3D Ewing sarcoma model that closely mimics the morphology, growth kinetics, and protein expression profile of human tumors. We observed that Ewing sarcoma cells cultured in porous 3D electrospun poly(ε-caprolactone) scaffolds not only were more resistant to traditional cytotoxic drugs than were cells in 2D monolayer culture but also exhibited remarkable differences in the expression pattern of the insulin-like growth factor-1 receptor/mammalian target of rapamycin pathway. This 3D model of the bone microenvironment may have broad applicability for mechanistic studies of bone sarcomas and exhibits the potential to augment preclinical evaluation of antineoplastic drug candidates for these malignancies.


Advanced Drug Delivery Reviews | 2014

3D tissue-engineered model of Ewing's sarcoma

Salah Eddine Lamhamedi-Cherradi; Marco Santoro; Vandhana Ramammoorthy; Brian A. Menegaz; Geoffrey Bartholomeusz; Lakesla R. Iles; Hesham M. Amin; J. Andrew Livingston; Antonios G. Mikos; Joseph A. Ludwig

Despite longstanding reliance upon monolayer culture for studying cancer cells, and numerous advantages from both a practical and experimental standpoint, a growing body of evidence suggests that more complex three-dimensional (3D) models are necessary to properly mimic many of the critical hallmarks associated with the oncogenesis, maintenance and spread of Ewings sarcoma (ES), the second most common pediatric bone tumor. And as clinicians increasingly turn to biologically-targeted therapies that exert their effects not only on the tumor cells themselves, but also on the surrounding extracellular matrix, it is especially important that preclinical models evolve in parallel to reliably measure antineoplastic effects and possible mechanisms of de novo and acquired drug resistance. Herein, we highlight a number of innovative methods used to fabricate biomimetic ES tumors, encompassing both the surrounding cellular milieu and the extracellular matrix (ECM), and suggest potential applications to advance our understanding of ES biology, preclinical drug testing, and personalized medicine.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Flow perfusion effects on three-dimensional culture and drug sensitivity of Ewing sarcoma

Marco Santoro; Salah Eddine Lamhamedi-Cherradi; Brian A. Menegaz; Joseph A. Ludwig; Antonios G. Mikos

Significance Most preclinical drug screening systems fail to predict whether a given drug candidate will succeed in clinical trials due to the fact that cells are plated on culture dishes devoid of signaling cues present in vivo. Among other factors, biomechanical stimulation strongly affects tumor behavior. Toward this end, we cultured Ewing sarcoma cells on 3D scaffolds within a flow perfusion bioreactor, which provides mechanical stimulation in the form of flow-derived shear stress. Cells exposed to flow perfusion produced more insulin-like growth factor-1 (IGF-1) ligand and showed shear stress-dependent sensitivity to IGF-1 receptor targeted drugs when compared with static conditions. Collectively, this research provides an improved method to study cancer cells ex vivo and could narrow the gap between preclinical and clinical drug activity. Three-dimensional tumor models accurately describe different aspects of the tumor microenvironment and are readily available for mechanistic studies of tumor biology and for drug screening. Nevertheless, these systems often overlook biomechanical stimulation, another fundamental driver of tumor progression. To address this issue, we cultured Ewing sarcoma (ES) cells on electrospun poly(ε-caprolactone) 3D scaffolds within a flow perfusion bioreactor. Flow-derived shear stress provided a physiologically relevant mechanical stimulation that significantly promoted insulin-like growth factor-1 (IGF1) production and elicited a superadditive release in the presence of exogenous IGF1. This finding is particularly relevant, given the central role of the IGF1/IGF-1 receptor (IGF-1R) pathway in ES tumorigenesis and as a promising clinical target. Additionally, flow perfusion enhanced in a rate-dependent manner the sensitivity of ES cells to IGF-1R inhibitor dalotuzumab (MK-0646) and showed shear stress-dependent resistance to the IGF-1R blockade. This study demonstrates shear stress-dependent ES cell sensitivity to dalotuzumab, highlighting the importance of biomechanical stimulation on ES-acquired drug resistance to IGF-1R inhibition. Furthermore, flow perfusion increased nutrient supply throughout the scaffold, enriching ES culture over static conditions. Our use of a tissue-engineered model, rather than human tumors or xenografts, enabled precise control of the forces experienced by ES cells, and therefore provided at least one explanation for the remarkable antineoplastic effects observed by some ES tumor patients from IGF-1R targeted therapies, in contrast to the lackluster effect observed in cells grown in conventional monolayer culture.


Journal of the National Cancer Institute | 2016

IGF-1R and mTOR Blockade: Novel Resistance Mechanisms and Synergistic Drug Combinations for Ewing Sarcoma

Salah Eddine Lamhamedi-Cherradi; Brian A. Menegaz; Vandhana Ramamoorthy; Deeksha Vishwamitra; Ying Wang; Rebecca L. Maywald; Adriana S. Buford; Izabela Fokt; Stanislaw Skora; Jing Wang; Aung Naing; Alexander J. Lazar; Eric Rohren; Najat C. Daw; Vivek Subbiah; Robert S. Benjamin; Ravin Ratan; Waldemar Priebe; Antonios G. Mikos; Hesham M. Amin; Joseph A. Ludwig

BACKGROUND Therapies cotargeting insulin-like growth factor receptor 1 (IGF-1R) and mammalian target of rapamycin (mTOR) have demonstrated remarkable, albeit short-lived, clinical responses in a subset of Ewing sarcoma (ES) patients. However, the mechanisms of resistance and applicable strategies for overcoming drug resistance to the IGF-1R/mTOR blockade are still undefined. METHODS To elucidate predominant mechanism(s) of acquired drug resistance while identifying synergistic drug combinations that improve clinical efficacy, we generated more than 18 ES cell lines resistant to IGF-1R- or mTOR-targeted therapy. Two small-molecule inhibitors of IGF-1R were chosen, NVP-ADW-742 (IGF-1R-selective) and OSI-906 (a dual IGF-1R/insulin receptor alpha [IR-α] inhibitor). Reverse-phase protein lysate arrays (RPPAs) revealed proteomic changes linked to IGF-1R/mTOR resistance, and selected proteins were validated in cell-based assays, xenografts, and within human clinical samples. All statistical tests were two-sided. RESULTS Novel mechanisms of resistance (MOR) emerged after dalotuzumab-, NVP-ADW-742-, and OSI-906-based targeting of IGF-1R. MOR to dalotuzumab included upregulation of IRS1, PI3K, and STAT3, as well as p38 MAPK, which was also induced by OSI-906. pEIF4E(Ser209), a key regulator of Cap-dependent translation, was induced in ridaforolimus-resistant ES cell lines. Unique drug combinations targeting IGF-1R and PI3K-alpha or Mnk and mTOR were synergistic in vivo and vitro (P < .001) as assessed respectively by Mantel-Cox and isobologram testing. CONCLUSIONS We discovered new druggable targets expressed by chemoresistant ES cells, xenografts, and relapsed human tumors. Joint suppression of these newfound targets, in concert with IGF-1R or mTOR blockade, should improve clinical outcomes.


Molecular Cancer Therapeutics | 2015

An Oral Formulation of YK-4-279: Preclinical Efficacy and Acquired Resistance Patterns in Ewing Sarcoma

Salah Eddine Lamhamedi-Cherradi; Brian A. Menegaz; Vandhana Ramamoorthy; Ramani Aiyer; Rebecca L. Maywald; Adrianna S. Buford; Dannette K. Doolittle; Kirk S. Culotta; James E. O'dorisio; Joseph A. Ludwig

Ewing sarcoma is a transcription factor–mediated pediatric bone tumor caused by a chromosomal translocation of the EWSR1 gene and one of several genes in the ETS family of transcription factors, typically FLI1 or ERG. Full activity of the resulting oncogenic fusion protein occurs only after binding RNA helicase A (RHA), and novel biologically targeted small molecules designed to interfere with that interaction have shown early promise in the preclinical setting. Herein, we demonstrate marked preclinical antineoplastic activity of an orally bioavailable formulation of YK-4-279 and identify mechanisms of acquired chemotherapy resistance that may be exploited to induce collateral sensitivity. Daily enteral administration of YK-4-279 led to significant delay in Ewing sarcoma tumor growth within a murine model. In advance of anticipated early-phase human clinical trials, we investigated both de novo and acquired mechanism(s) by which Ewing sarcoma cells evade YK-4-279–mediated cell death. Drug-resistant clones, formed by chronic in vitro exposure to steadily increased levels of YK-4-279, overexpressed c-Kit, cyclin D1, pStat3(Y705), and PKC isoforms. Interestingly, cross-resistance to imatinib and enzastaurin (selective inhibitors of c-Kit and PKC-β, respectively), was observed and the use of YK-4-279 with enzastaurin in vitro led to marked drug synergy, suggesting a potential role for combination therapies in the future. By advancing an oral formulation of YK-4-279 and identifying prominent mechanisms of resistance, this preclinical research takes us one step closer to a shared goal of curing adolescents and young adults afflicted by Ewing sarcoma. Mol Cancer Ther; 14(7); 1591–604. ©2015 AACR.


Tissue Engineering Part A | 2016

Modeling Stroma-Induced Drug Resistance in a Tissue-Engineered Tumor Model of Ewing Sarcoma

Marco Santoro; Brian A. Menegaz; Salah Eddine Lamhamedi-Cherradi; Eric R. Molina; Danielle Wu; Waldemar Priebe; Joseph A. Ludwig; Antonios G. Mikos

Three-dimensional (3D) tumor models are gaining traction in the research community given their capacity to mimic aspects of the tumor microenvironment absent in monolayer systems. In particular, the ability to spatiotemporally control cell placement within ex vivo 3D systems has enabled the study of tumor-stroma interactions. Furthermore, by regulating biomechanical stimuli, one can reveal how biophysical cues affect stromal cell phenotype and how their phenotype impacts tumor drug sensitivity. Both tumor architecture and shear force have profound effects on Ewing sarcoma (ES) cell behavior and are known to elicit ligand-mediated activation of the insulin-like growth factor-1 receptor (IGF-1R), thereby mediating resistance of ES cells to IGF-1R inhibitors. Here, we demonstrate that these same biophysical cues-modeled by coculturing ES cells and mesenchymal stem cells (MSCs) in 3D scaffolds within a flow perfusion bioreactor-activate interleukin-6 and transcription factor Stat3. Critically, an active Stat3 pathway drastically alters the equilibrium of IGF-1R-targeted ligands (IGF-1) and antagonists (IGFBP-3) secreted by MSCs. To elucidate how this might promote ES tumor growth under physiological shear-stress conditions, ES cells and MSCs were co-cultured by using a flow perfusion bioreactor at varying ratios that simulate a wide range of native MSC abundance. Our results indicate that ES cells and MSCs stimulate each others growth. Co-targeting IGF-1R and Stat3 enhanced antineoplastic activity over monotherapy treatment. Although this discovery requires prospective clinical validation in patients, it reveals the power of employing a more physiological tissue-engineered 3D tumor model to elucidate how tumor cells co-opt stromal cells to acquire drug resistance.


Oncologist | 2017

Clinical Activity of Pazopanib in Patients with Advanced Desmoplastic Small Round Cell Tumor

Brian A. Menegaz; Branko Cuglievan; Jalen Benson; Pamela Camacho; Salah Eddine Lamhamedi-Cherradi; Cheuk Hong Leung; Carla L. Warneke; Winston W. Huh; Vivek Subbiah; Robert S. Benjamin; Shreyaskumar Patel; Najat C. Daw; Andrea Hayes-Jordan; Joseph A. Ludwig

Desmoplastic small round cell tumor (DSRCT) is an aggressive, usually incurable, soft tissue sarcoma subtype that presents with diffuse abdominal sarcomatosis in adolescents and young adults. This article presents the largest retrospective analysis conducted to date of patients with DSRCT who were treated with an anti‐angiogenic therapy.


Neoplasia | 2018

Efficacy of ONC201 in Desmoplastic Small Round Cell Tumor

Andrea Hayes-Jordan; Xiao Ma; Brian A. Menegaz; Salah Eddine Lamhamedi-Cherradi; Charles Kingsley; Jalen Benson; Pamela Camacho; Joseph A. Ludwig; Cynthia R. Lockworth; Gloria E. Garcia; Suzanne Craig

Desmoplastic Small Round Cell Tumor (DSRCT) is a rare sarcoma tumor of adolescence and young adulthood, which harbors a recurrent chromosomal translocation between the Ewing’s sarcoma gene (EWSR1) and the Wilms’ tumor suppressor gene (WT1). Patients usually develop multiple abdominal tumors with liver and lymph node metastasis developing later. Survival is poor using a multimodal therapy that includes chemotherapy, radiation and surgical resection, new therapies are needed for better management of DSRCT. Triggering cell apoptosis is the scientific rationale of many cancer therapies. Here, we characterized for the first time the expression of pro-apoptotic receptors, tumor necrosis-related apoptosis-inducing ligand receptors (TRAILR1-4) within an established human DSRCT cell line and clinical samples. The molecular induction of TRAIL-mediated apoptosis using agonistic small molecule, ONC201 in vitro cell-based proliferation assay and in vivo novel orthotopic xenograft animal models of DSRCT, was able to inhibit cell proliferation that was associated with caspase activation, and tumor growth, indicating that a cell-based delivery of an apoptosis-inducing factor could be relevant therapeutic agent to control DSRCT.


Clinical Cancer Research | 2018

Multimodality Treatment of Desmoplastic small round cell tumor: Chemotherapy and Complete Cytoreductive Surgery Improve Patient Survival

Vivek Subbiah; Salah-Eddine Lamhamedi-Cherradi; Branko Cuglievan; Brian A. Menegaz; Pamela Camacho; Winston W. Huh; Vandhana Ramamoorthy; Peter M. Anderson; Raphael E. Pollock; Dina Lev; Wei Qiao; Mary Frances McAleer; Robert S. Benjamin; Shreyaskumar Patel; Cynthia E. Herzog; Najat C. Daw; Barry W. Feig; Alexander J. Lazar; Andrea Hayes-Jordan; Joseph A. Ludwig

Purpose: Desmoplastic small round cell tumor (DSRCT), which harbors EWSR1-WT1 t(11;22)(p13:q12) chromosomal translocation, is an aggressive malignancy that typically presents as intra-abdominal sarcomatosis in young males. Given its rarity, optimal treatment has not been defined. Experimental Design: We conducted a retrospective study of 187 patients with DSRCT treated at MD Anderson Cancer Center over 2 decades. Univariate and multivariate regression analyses were performed. We determined whether chemotherapy, complete cytoreductive surgery (CCS), hyperthermic intraperitoneal cisplatin (HIPEC), and/or whole abdominal radiation (WART) improve overall survival (OS) in patients with DSRCT. Critically, because our institutional practice limits HIPEC and WART to patients with less extensive, potentially resectable disease that had benefited from neoadjuvant chemotherapy, a time-variant analysis was performed to evaluate those adjunct treatment modalities. Results: The pre-2003 5-year OS rate of 5% has substantially improved to 25% with the advent of newer chemotherapies and better surgical and radiotherapy techniques (HR, 0.47; 95% CI, 0.29–0.75). Chemotherapy response (log rank P = 0.004) and CCS (log rank P < 0.0001) were associated with improved survival. Although WART and HIPEC lacked statistical significance, our study was not powered to detect their potential impact upon OS. Conclusions: Improved 3- and 5-year OS were observed following multidisciplinary treatment that includes Ewing sarcoma (ES)-based chemotherapy and complete tumor cytoreductive surgery, but few if any patients are cured. Prospective randomized studies will be required to prove whether HIPEC or WART are important. In the meantime, chemotherapy and CCS remain the cornerstone of treatment and provide a solid foundation to evaluate new biologically targeted therapies. Clin Cancer Res; 24(19); 4865–73. ©2018 AACR.


Pediatric Blood & Cancer | 2017

A pediatric patient with glycogen storage disease type IA and Castleman disease.

Branko Cuglievan; Brian A. Menegaz; April DePombo; Ajaykumar C. Morani; Roberto N. Miranda; Michael Rytting

Patients with glycogen storage disease type IA (GSD-IA) have a higher than average risk of developing various abdominal tumors .1–5 The pathophysiology of these tumors has been related to the underlying metabolic disorder secondary to mutations in the G6PC gene.3,6 Castleman disease is a rare lymphoproliferative disorder that has never been reported in association with GSD. A 15-year-old male with GSD-IA diagnosed at 5 months of age through liver biopsy presented to his pediatrician for routine assessment for hepatosplenomegaly. Abdominal ultrasonography revealed a paraspinal, partially cystic mass measuring 4.4 × 3.4 × 2.5 cm. Abdominal computed tomography (CT) showed a lesion displacing the right adrenal gland inferiorly. Attenuation and washout values were not characteristic of an adenoma (Figure 1A). Complete blood count, electrolytes, tests for hormone levels and viral diseases were negative. The mass was surgically excised. Histopathologic examination demonstrated numerous lymphoid follicles surrounded by a dense infiltrate of small lymphocytes and scattered mature plasma cells (Figure 1B). The follicles were poorly defined and composed of small lymphocytes in a focal concentric arrangement around germinal centers (Figure 1C). Most germinal centers were involuted and partially replaced by follicular dendritic cells admixed with hyalinized vessels (Figure 1D). In immunohistochemical studies, most lymphoid follicles stained positive for CD20, whilemost interfollicular lymphocyteswere highlighted withCD3. The anti-CD31 antibody highlighted themarkedly increased vasculature in interfollicular areas as well as the small vessels within the germinal centers. A diagnosis of the hyaline-vascular variant of Castleman disease was rendered. Castleman disease is a reactive process characterized by benign hyperplastic lymph nodes, follicular hyperplasia with involuted germinal centers, polyclonal plasma cells, and capillary proliferation in the interfollicular areas.7 Though the etiopathology is unknown, the generation of interleukin (IL)-6 in germinal centers of hyperplastic lymph nodes, resulting in inductionofB-cell differentiation, seems tobe implicated in the etiopathogenesis of the disease.8 Castleman disease has not been reported in association with GSD; however, this group of metabolic diseases has been linked with hepatocellular adenomas, hepatoblastomas, and hepatocellular carcinomas.1–5 The underlying pathophysiology of cell proliferation in these tumors remains poorly understood; nevertheless, the metabolic amendment found in gluconeogenesis and glycogenolysis due to the absence of glucose-6-phosphatase has been proposed as a triggering mechanism of cellular overgrowth.2,6

Collaboration


Dive into the Brian A. Menegaz's collaboration.

Top Co-Authors

Avatar

Joseph A. Ludwig

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Salah Eddine Lamhamedi-Cherradi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Branko Cuglievan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Vandhana Ramamoorthy

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander J. Lazar

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Andrea Hayes-Jordan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hesham M. Amin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Najat C. Daw

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge