Spencer Rosario
Roswell Park Cancer Institute
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Spencer Rosario.
Science | 2017
Sheng Yu Ku; Spencer Rosario; Yanqing Wang; Ping Mu; Mukund Seshadri; Zachary W. Goodrich; Maxwell M. Goodrich; David P. Labbé; Eduardo Cortes Gomez; Jianmin Wang; Henry W. Long; Bo Xu; Myles Brown; Massimo Loda; Charles L. Sawyers; Leigh Ellis; David W. Goodrich
Evading cancer drugs by identity fraud Prostate cancer growth is fueled by male hormones called androgens. Drugs targeting the androgen receptor (AR) are initially efficacious, but most tumors eventually become resistant (see the Perspective by Kelly and Balk). Mu et al. found that prostate cancer cells escaped the effects of androgen deprivation therapy through a change in lineage identity. Functional loss of the tumor suppressors TP53 and RB1 promoted a shift from AR-dependent luminal epithelial cells to AR-independent basal-like cells. In related work, Ku et al. found that prostate cancer metastasis, lineage switching, and drug resistance were driven by the combined loss of the same tumor suppressors and were accompanied by increased expression of the epigenetic regulator Ezh2. Ezh2 inhibitors reversed the lineage switch and restored sensitivity to androgen deprivation therapy in experimental models. Science, this issue p. 84, p. 78; see also p. 29 Prostate cancer cells escape androgen deprivation therapy by morphing into a cell type that does not require androgens. Prostate cancer relapsing from antiandrogen therapies can exhibit variant histology with altered lineage marker expression, suggesting that lineage plasticity facilitates therapeutic resistance. The mechanisms underlying prostate cancer lineage plasticity are incompletely understood. Studying mouse models, we demonstrate that Rb1 loss facilitates lineage plasticity and metastasis of prostate adenocarcinoma initiated by Pten mutation. Additional loss of Trp53 causes resistance to antiandrogen therapy. Gene expression profiling indicates that mouse tumors resemble human prostate cancer neuroendocrine variants; both mouse and human tumors exhibit increased expression of epigenetic reprogramming factors such as Ezh2 and Sox2. Clinically relevant Ezh2 inhibitors restore androgen receptor expression and sensitivity to antiandrogen therapy. These findings uncover genetic mutations that enable prostate cancer progression; identify mouse models for studying prostate cancer lineage plasticity; and suggest an epigenetic approach for extending clinical responses to antiandrogen therapy.
Clinical Cancer Research | 2017
David P. Labbé; Christopher Sweeney; Myles Brown; Phillip Galbo; Spencer Rosario; Kristine M. Wadosky; Sheng Yu Ku; Martin Sjöström; Mohammed Alshalalfa; Nicholas Erho; Elai Davicioni; R. Jeffrey Karnes; Edward M. Schaeffer; Robert B. Jenkins; Robert B. Den; Ashley E. Ross; Michaela Bowden; Ying Huang; Kathryn P. Gray; Felix Y. Feng; Daniel E. Spratt; David W. Goodrich; Kevin H. Eng; Leigh Ellis
Purpose: Current clinical parameters do not stratify indolent from aggressive prostate cancer. Aggressive prostate cancer, defined by the progression from localized disease to metastasis, is responsible for the majority of prostate cancer–associated mortality. Recent gene expression profiling has proven successful in predicting the outcome of prostate cancer patients; however, they have yet to provide targeted therapy approaches that could inhibit a patients progression to metastatic disease. Experimental Design: We have interrogated a total of seven primary prostate cancer cohorts (n = 1,900), two metastatic castration-resistant prostate cancer datasets (n = 293), and one prospective cohort (n = 1,385) to assess the impact of TOP2A and EZH2 expression on prostate cancer cellular program and patient outcomes. We also performed IHC staining for TOP2A and EZH2 in a cohort of primary prostate cancer patients (n = 89) with known outcome. Finally, we explored the therapeutic potential of a combination therapy targeting both TOP2A and EZH2 using novel prostate cancer–derived murine cell lines. Results: We demonstrate by genome-wide analysis of independent primary and metastatic prostate cancer datasets that concurrent TOP2A and EZH2 mRNA and protein upregulation selected for a subgroup of primary and metastatic patients with more aggressive disease and notable overlap of genes involved in mitotic regulation. Importantly, TOP2A and EZH2 in prostate cancer cells act as key driving oncogenes, a fact highlighted by sensitivity to combination-targeted therapy. Conclusions: Overall, our data support further assessment of TOP2A and EZH2 as biomarkers for early identification of patients with increased metastatic potential that may benefit from adjuvant or neoadjuvant targeted therapy approaches. Clin Cancer Res; 23(22); 7072–83. ©2017 AACR.
Oncotarget | 2017
Hayley C. Affronti; Mark D. Long; Spencer Rosario; Bryan M. Gillard; Ellen Karasik; Christoph S. Boerlin; Anthony J. Pellerite; Barbara A. Foster; Kristopher Attwood; Roberto Pili; John Wilton; Moray J. Campbell; Dominic J. Smiraglia
Folate impacts the genome and epigenome by feeding into one-carbon metabolism to produce critical metabolites, deoxythymidine monophosphate and s-adenosylmethionine. The impact of folate exposure and intervention timing on cancer progression remains controversial. Due to polyamine metabolism’s extraordinary biosynthetic flux in prostate cancer (CaP) we demonstrated androgen stimulated CaP is susceptible to dietary folate deficiency. We hypothesized dietary folate levels may also affect castration recurrent CaP. We used the CWR22 human xenograft model which recurs following androgen withdrawal. Engrafted mice were fed a folate depleted or supplemented diet beginning at androgen withdrawal, or prior to xenograft implantation. Both folate depletion and supplementation at the time of withdrawal significantly decreased recurrence incidence. Folate supplementation prior to xenograft implantation increased time to recurrence, suggesting a protective role. By contrast, folate depleted recurrent tumors exhibited transcriptional adaptive responses that maintained high polyamine levels at the expense of increased DNA damage and DNA methylation alterations. Mining of publically available data demonstrated folate related pathways are exceptionally dysregulated in human CaP, which correlated with decreased time to biochemical recurrence. These findings highlight the potential for novel therapeutic interventions that target these metabolic pathways in CaP and provide a rationale to apply such strategies alongside androgen withdrawal.
Cell Death and Disease | 2015
Elena Lasorsa; M Smonksey; Jason Kirk; Spencer Rosario; F J Hernandez-Ilizaliturri; Leigh Ellis
Inhibitors of the bromodomain and extraterminal domain family (BETI) have recently entered phase I clinical trials. In patients with advanced leukemia’s, potent antileukemia activity was displayed with minimum dose-limiting toxicity. In preclinical models of hematological malignancies, including aggressive B-cell lymphomas, BETI induced cell-cycle arrest and apoptosis. However, the underlying cell death mechanisms are still not well understood. Dissecting the mechanisms required by BETI to mediate cell death would provide strong direction on how to best utilize BETI to treat patients with aggressive hematological malignancies. Herein, we provide understanding of the molecular mechanisms underlying BETI-mediated cell death using I-BET762. Induction of cell death occurred in primary murine and human B-cell lymphomas through apoptosis. Genetic dissection using Eμ-myc B-cell lymphoma compound mutants demonstrated that I-BET762-induced apoptosis does not require the p53 pathway. Furthermore, deletion of Apaf1, and thus the absence of a functional apoptosome, is associated with a delayed drug response but do not provide long-term resistance. Prolonged treatment of this model in fact fails to suppress the therapeutic efficacy of the drug and is associated with biochemical features of autophagy. However, lack of mitochondrial permeability completely inhibited I-BET762-mediated tumor cell death, indicating mitochondrial damage as key events for its activity. Combination of I-BET762 with BH3-only mimetics ABT-263 or obatoclax, restored sensitivity to I-BET762 lymphoma killing; however, success was determined by expression of Bcl-2 family antiapoptotic proteins. Our study provides critical insight for clinical decisions regarding the appropriate strategy for using BETI as a single agent or in combination to treat patients with aggressive B-cell lymphomas.
Oncogene | 2018
Mark D. Long; Prashant K. Singh; James R. Russell; Gerard Llimos; Spencer Rosario; Abbas Rizvi; Patrick R. van den Berg; Jason Kirk; Lara E. Sucheston-Campbell; Dominic J. Smiraglia; Moray J. Campbell
Expression levels of retinoic acid receptor gamma (NR1B3/RARG, encodes RARγ) are commonly reduced in prostate cancer (PCa). Therefore, we sought to establish the cellular and gene regulatory consequences of reduced RARγ expression, and determine RARγ regulatory mechanisms. RARG shRNA approaches in non-malignant (RWPE-1 and HPr1-AR) and malignant (LNCaP) prostate models revealed that reducing RARγ levels, rather than adding exogenous retinoid ligand, had the greatest impact on prostate cell viability and gene expression. ChIP-Seq defined the RARγ cistrome, which was significantly enriched at active enhancers associated with AR binding sites. Reflecting a significant genomic role for RARγ to regulate androgen signaling, RARγ knockdown in HPr1-AR cells significantly regulated the magnitude of the AR transcriptome. RARγ downregulation was explained by increased miR-96 in PCa cell and mouse models, and TCGA PCa cohorts. Biochemical approaches confirmed that miR-96 directly regulated RARγ expression and function. Capture of the miR-96 targetome by biotin-miR-96 identified that RARγ and a number of RARγ interacting co-factors including TACC1 were all targeted by miR-96, and expression of these genes were prominently altered, positively and negatively, in the TCGA-PRAD cohort. Differential gene expression analyses between tumors in the TCGA-PRAD cohort with lower quartile expression levels of RARG and TACC1 and upper quartile miR-96, compared to the reverse, identified a gene network including several RARγ target genes (e.g., SOX15) that significantly associated with worse disease-free survival (hazard ratio 2.23, 95% CI 1.58 to 2.88, p = 0.015). In summary, miR-96 targets a RARγ network to govern AR signaling, PCa progression and disease outcome.
Journal of Clinical Investigation | 2018
Anna Bianchi-Smiraglia; Archis Bagati; Emily E. Fink; Hayley C. Affronti; Brittany C. Lipchick; Sudha Moparthy; Mark D. Long; Spencer Rosario; Shivana M. Lightman; Kalyana Moparthy; David W. Wolff; Dong Hyun Yun; Zhannan Han; Anthony Polechetti; Matthew V. Roll; Ilya Gitlin; Katerina I. Leonova; Aryn M. Rowsam; Eugene S. Kandel; Andrei V. Gudkov; P. Leif Bergsagel; Kelvin P. Lee; Dominic J. Smiraglia; Mikhail A. Nikiforov
Polyamine inhibition for cancer therapy is, conceptually, an attractive approach but has yet to meet success in the clinical setting. The aryl hydrocarbon receptor (AHR) is the central transcriptional regulator of the xenobiotic response. Our study revealed that AHR also positively regulates intracellular polyamine production via direct transcriptional activation of 2 genes, ODC1 and AZIN1, which are involved in polyamine biosynthesis and control, respectively. In patients with multiple myeloma (MM), AHR levels were inversely correlated with survival, suggesting that AHR inhibition may be beneficial for the treatment of this disease. We identified clofazimine (CLF), an FDA-approved anti-leprosy drug, as a potent AHR antagonist and a suppressor of polyamine biosynthesis. Experiments in a transgenic model of MM (Vk*Myc mice) and in immunocompromised mice bearing MM cell xenografts revealed high efficacy of CLF comparable to that of bortezomib, a first-in-class proteasome inhibitor used for the treatment of MM. This study identifies a previously unrecognized regulatory axis between AHR and polyamine metabolism and reveals CLF as an inhibitor of AHR and a potentially clinically relevant anti-MM agent.
bioRxiv | 2018
Spencer Rosario; Mark D. Long; Hayley C. Affronti; Aryn M. Rowsam; Kevin H. Eng; Dominic J. Smiraglia
Understanding the levels of metabolic dysregulation in different disease settings is vital for the safe and effective incorporation of metabolism-targeted therapeutics in the clinic. Using transcriptomic data from 10,704 tumor and normal samples from The Cancer Genome Atlas, across 26 disease sites, we developed a novel bioinformatics pipeline that distinguishes tumor from normal tissues, based on differential gene expression for 114 metabolic pathways. This pathway dysregulation was confirmed in separate patient populations, further demonstrating the robustness of this approach. A bootstrapping simulation was then applied to assess whether these alterations were biologically meaningful, rather than expected by chance. We provide distinct examples of the types of analysis that can be accomplished with this tool to understand cancer specific metabolic dysregulation, highlighting novel pathways of interest in both common and rare disease sites. Utilizing a pathway mapping approach to understand patterns of metabolic flux, differential drug sensitivity, can accurately be predicted. Further, the identification of Master Metabolic Transcriptional Regulators, whose expression was highly correlated with pathway gene expression, explains why metabolic differences exist in different disease sites. We demonstrate these also have the ability to segregate patient populations and predict responders to different metabolism-targeted therapeutics.
Cancer Research | 2018
Mark D. Long; Prashant K. Singh; Gerard Llimos; Spencer Rosario; Dominic J. Smiraglia; Moray J. Campbell
The nuclear receptor corepressors (NCORs) serve as critical mediators between transcription factors and epigenetic regulators, and thus their functions are critical in the maintenance of epigenetic and transcriptional states. As such, NCORs have important developmental and homeostatic roles, and disruptions of their functions are implicated in various cancers. In hormonally driven cancers, such as breast and prostate cancer (PCa), NCORs are implicated in de novo and acquired changes to steroid hormone signaling and therapeutic resistance. Supporting the importance of NCOR function in the prostate, inactivating genomic aberrations to NCORs have been identified in androgen deprivation therapy (ADT) resistant disease. However, to date, the global functions of NCORs in prostate cancer cells and their overall contributions to PCa progression remain enigmatic. To characterize the global functions of NCOR2/SMRT in prostate cancer we have undertaken efforts to map the NCOR2 dependent transcriptome (RNA-seq), miRnome (miRNA-seq), methylome (EPIC methylation array) and cistrome (ChIP-seq) in both androgen-sensitive (LNCaP) and ADT-resistant (LNCaP-C42) prostate cancer cells upon shRNA targeted knockdown. NCOR2 loss in LNCaP-C42 cells revealed a striking degree of global hypermethylation (87,078 CpGs > 10% gain, adj.pval 10% loss). Hypermethylation was largely exclusive of TSS loci and CpG islands, occurring at distal regions. Conversely, the NCOR2 associated transcriptome was more divergent in its expression changes by comparable criteria (1,491 upregulated, 1,195 downregulated). Similar patterns were observed in LNCaP cells, albeit to a lesser extent, suggesting broader regulatory roles in ADT resistant cells. NCOR2 knockdown deregulated a number of miRNA including let-7e and miR-200a with target binding motifs significantly enriched among the NCOR2 regulated transcriptome, and several of these miRNA stratified survival in PCa patient data. Furthermore, NCOR2 binding at candidate loci distinguished normal from malignant prostate cells, and global binding profiles revealed significant overlap with androgen receptor and pioneering factor FOXA1. To examine the effect of NCOR2 loss in the context of androgen signaling, similar experimental conditions were performed in the presence of dihydrotestosterone (DHT). NCOR2 control of androgen sensitivity was complex, resulting in amplified signaling of some classic AR target genes (KLK3, HERC3) while dampening others (TMPRSS2, TIPARP). Ongoing data integration efforts will dissect novel NCOR2 methylome-cistrome-transcriptome relationships within and across models to define target gene networks that are epigenetically governed through NCOR2 function. To determine whether NCOR2 loss alters PCa progression we have utilized stable introduction of NCOR2 targeting shRNA in the CWR22 xenograft model that simulates androgen-dependent primary growth, regression upon ADT, and subsequent recurrence at established proportions. NCOR2 loss had no effect on primary tumor growth rate or size of tumor at ADT. However, shNCOR2 expressing tumors had significantly reduced regression in response to ADT, and furthermore at 180 days post injection, a significantly higher proportion of shNCOR2 expressing tumors (17/49) had reached recurrence compared to shCTL tumors (7/46). In total, these findings place NCOR2 as an important epigenetic regulator in prostate cancer cells, the dysfunction of which contributes to PCa progression. Citation Format: Mark D. Long, Prashant K. Singh, Gerard Llimos, Spencer Rosario, Dominic J. Smiraglia, Moray J. Campbell. Characterizing the global function of NCOR2 in prostate cancer cells and its contribution to PCa progression [abstract]. In: Proceedings of the AACR Special Conference: Prostate Cancer: Advances in Basic, Translational, and Clinical Research; 2017 Dec 2-5; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(16 Suppl):Abstract nr B033.
bioRxiv | 2017
Mark D. Long; Prashant K. Singh; James R. Russell; Gerard Llimos; Spencer Rosario; Abbas Rizvi; Henry G. Withers; Patrick R. van den Berg; Jason Kirk; Lara E. Sucheston-Campbell; Dominic J. Smiraglia; Moray J. Campbell
Expression levels of retinoic acid receptor gamma (NR1B3/RARG, encodes RARγ), are commonly reduced in prostate cancer (PCa). Therefore we sought to establish the cellular and gene regulatory consequences of reduced RARγ expression, and determine RARγ regulatory mechanisms. RARG shRNA approaches in non-malignant (RWPE-1 and HPr1-AR) and malignant (LNCaP) prostate models revealed that reducing RARγ levels, rather than adding exogenous retinoid ligand, had the greatest impact on prostate cell viability and gene expression. ChIP-Seq defined the RARγ cistrome which was significantly enriched at active enhancers associated with AR binding sites. Reflecting a significant genomic role for RARγ to regulate androgen signaling, RARγ knockdown in HPr1-AR cells significantly regulated the magnitude of the AR transcriptome. RARγ down-regulation was explained by increased miR-96 in PCa cell and mouse models, and TCGA PCa cohorts. Biochemical approaches confirmed that miR-96 directly regulated RARγ expression and function Capture of the miR-96 targetome by biotin-miR96 identified that RARγ and a number of RARγ interacting co-factors including TACC1 were all targeted by miR-96, and expression of these genes were prominently altered, positively and negatively, in the TCGA-PRAD cohort. Differential gene expression analyses between tumors in the TCGA-PRAD cohort with lower quartile expression levels of RARG and TACC1 and upper quartile miR-96, compared to the reverse, identified a gene network including several RARγ target genes (e.g. SOX15) that significantly associated with worse disease free survival (hazard ratio 2.23, 95% CI 1.58 to 2.88, p=0.015). In summary, miR-96 targets a RARγ network to govern AR signaling, PCa progression and disease outcome. Conflict of interest: The authors certify that they have NO affiliations with or involvement in any organization or entity with any financial interest (such as honoraria; educational grants; participation in speakers’ bureaus; membership, employment, consultancies, stock ownership, or other equity interest; and expert testimony or patent-licensing arrangements), or non-financial interest (such as personal or professional relationships, affiliations, knowledge or beliefs) in the subject matter or materials discussed in this manuscript. FUNDING LESC acknowledges support, in part, of Roswell Park Comprehensive Cancer Center-University of Pittsburg Cancer Institute Ovarian Cancer Specialized Program of Research Excellence National Institutes of Health [P50CA159981-01A1]. MDL acknowledges support of Molecular Pharmacology and Experimental Therapeutics NRSA T32 program [T32CA009072] held at Roswell Park Comprehensive Cancer Center. MJC and DJS acknowledges support in part from the Prostate program of the Department of Defense Congressionally Directed Medical Research Programs [W81XWH-14-1-0608, W81XWH-11-2-0033] and the National Cancer Institute (NCI) grant P30CA016056 involving the use of Roswell Park Comprehensive Cancer Center’s Genomic Shared Resource. MJC, GL, AR, HW and PvdB acknowledges support from the European Union-United States Atlantis Program [P116J090011]. MJC and LESC acknowledge support from the National Cancer Institute (NCI) grant P30CA016056 involving the use of OSUCCC The James, CCSG P30CA016058
Oncoscience | 2016
Matthew T. Smonskey; Elena Lasorsa; Spencer Rosario; Jason Kirk; Francisco J. Hernandez-Ilizaliturri; Leigh Ellis