33 results on '"Christopher Fiore"'
Search Results
2. Antitumor synergy with SY-1425, a selective RARα agonist, and hypomethylating agents in retinoic acid receptor pathway activated models of acute myeloid leukemia
- Author
-
Michael R. McKeown, Liv Johannessen, Emily Lee, Christopher Fiore, and Emmanuelle di Tomaso
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2019
- Full Text
- View/download PDF
3. Targeting RARA overexpression with tamibarotene, a potent and selective RARα agonist, is a novel approach in AML
- Author
-
Stéphane de Botton, Thomas Cluzeau, Carlos Vigil, Rachel J. Cook, Philippe Rousselot, David A. Rizzieri, Jane L. Liesveld, Pierre Fenaux, Thorsten Braun, Anne Banos, Joseph G. Jurcic, Mikkael A. Sekeres, Michael R. Savona, Gail J. Roboz, Dale Bixby, Kate Madigan, Angela Volkert, Kristin Stephens, Qing Kang-Fortner, Kristen Baker, Sofia Paul, Michael McKeown, John Carulli, Matthew Eaton, Graeme Hodgson, Christopher Fiore, Michael J. Kelly, David A. Roth, and Eytan M. Stein
- Subjects
Hematology - Abstract
A superenhancer at the retinoic acid receptor alpha (RARA) gene is associated with RARA mRNA overexpression in ∼30% of non-acute promyelocytic leukemia acute myeloid leukemia (AML) and in ∼50% of myelodysplastic syndromes (MDS). RARA overexpression is an actionable target for treatment with tamibarotene, an oral potent and selective RARα agonist. Sensitivity to the RARα agonist tamibarotene was demonstrated in RARA-high but not RARA-low preclinical AML models. The combination of oral tamibarotene plus azacitidine was evaluated in a phase 2 clinical study in 51 newly diagnosed unfit patients with AML identified as RARA-positive (n = 22) or RARA-negative (n = 29) for RARA mRNA overexpression in peripheral blasts using a blood-based biomarker test. In 18 response-evaluable RARA-positive patients, complete remission (CR)/CR with incomplete hematologic recovery rate was 61%, CR rate was 50%, and time to initial composite CR was rapid at 1.2 months. Transfusion independence was attained by 72% of RARA-positive patients. In contrast, 28 response-evaluable RARA-negative patients had response rates that were consistent with azacitidine monotherapy. Tamibarotene in combination with azacitidine was well tolerated. The majority of nonhematologic adverse events were low grade and hematologic adverse events were comparable to single-agent azacitidine, demonstrating that there was no additional myelosuppression when tamibarotene was combined with azacitidine. These results support further evaluation of tamibarotene-based treatment strategies in patients with AML or MDS with RARA overexpression to provide a targeted approach with the goal of improving patient outcomes. This trial was registered at www.clinicaltrials.gov as #NCT02807558.
- Published
- 2023
4. ΔNp63/p73 drive metastatic colonization by controlling a regenerative epithelial stem cell program in quasi-mesenchymal cancer stem cells
- Author
-
Arthur W. Lambert, Christopher Fiore, Yogesh Chutake, Elisha R. Verhaar, Patrick C. Strasser, Mei Wei Chen, Daneyal Farouq, Sunny Das, Xin Li, Elinor Ng Eaton, Yun Zhang, Joana Liu Donaher, Ian Engstrom, Ferenc Reinhardt, Bingbing Yuan, Sumeet Gupta, Bruce Wollison, Matthew Eaton, Brian Bierie, John Carulli, Eric R. Olson, Matthew G. Guenther, and Robert A. Weinberg
- Subjects
Epithelial-Mesenchymal Transition ,Cell Line, Tumor ,Neoplasms ,Neoplastic Stem Cells ,Humans ,Mesenchymal Stem Cells ,Cell Biology ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology ,Signal Transduction ,Developmental Biology - Abstract
Cancer stem cells (CSCs) may serve as the cellular seeds of tumor recurrence and metastasis, and they can be generated via epithelial-mesenchymal transitions (EMTs). Isolating pure populations of CSCs is difficult because EMT programs generate multiple alternative cell states, and phenotypic plasticity permits frequent interconversions between these states. Here, we used cell-surface expression of integrin β4 (ITGB4) to isolate highly enriched populations of human breast CSCs, and we identified the gene regulatory network operating in ITGB4
- Published
- 2022
5. Antitumor synergy with SY-1425, a selective RARα agonist, and hypomethylating agents in retinoic acid receptor pathway activated models of acute myeloid leukemia
- Author
-
Emily Lee, Michael R. McKeown, Christopher Fiore, Emmanuelle di Tomaso, and Liv Johannessen
- Subjects
Agonist ,Tetrahydronaphthalenes ,business.industry ,medicine.drug_class ,Retinoic Acid Receptor alpha ,Myeloid leukemia ,Hematology ,medicine.disease ,Benzoates ,Xenograft Model Antitumor Assays ,Neoplasm Proteins ,Mice ,Leukemia ,Retinoic acid receptor ,Leukemia, Promyelocytic, Acute ,Cell culture ,Cell Line, Tumor ,Cancer research ,Animals ,Humans ,Medicine ,Online Only Articles ,business ,Signal Transduction - Published
- 2018
6. Functional cis-regulatory modules encoded by mouse-specific endogenous retrovirus
- Author
-
Christopher Fiore, Tammy Ray, Barak A. Cohen, Brett B Maricque, Erica C. Pehrsson, Mayank N. K. Choudhary, Ting Wang, Vasavi Sundaram, Ava Hughes, Manishi Pandey, Yujie Chen, Xiaoyun Xing, Methma Udawatta, Duc Ngo, and Asia Paguntalan
- Subjects
0301 basic medicine ,Transposable element ,Science ,General Physics and Astronomy ,Endogenous retrovirus ,Computational biology ,Biology ,Genome ,General Biochemistry, Genetics and Molecular Biology ,Article ,Gene Expression Regulation, Enzymologic ,Evolution, Molecular ,03 medical and health sciences ,Mice ,Animals ,Enhancer ,Gene ,Transcription factor ,Embryonic Stem Cells ,Cis-regulatory module ,Genetics ,Multidisciplinary ,Terminal Repeat Sequences ,food and beverages ,General Chemistry ,3. Good health ,030104 developmental biology ,Regulatory sequence ,DNA Transposable Elements ,Transcription Factors - Abstract
Cis-regulatory modules contain multiple transcription factor (TF)-binding sites and integrate the effects of each TF to control gene expression in specific cellular contexts. Transposable elements (TEs) are uniquely equipped to deposit their regulatory sequences across a genome, which could also contain cis-regulatory modules that coordinate the control of multiple genes with the same regulatory logic. We provide the first evidence of mouse-specific TEs that encode a module of TF-binding sites in mouse embryonic stem cells (ESCs). The majority (77%) of the individual TEs tested exhibited enhancer activity in mouse ESCs. By mutating individual TF-binding sites within the TE, we identified a module of TF-binding motifs that cooperatively enhanced gene expression. Interestingly, we also observed the same motif module in the in silico constructed ancestral TE that also acted cooperatively to enhance gene expression. Our results suggest that ancestral TE insertions might have brought in cis-regulatory modules into the mouse genome., The gene-battery model posits transposable elements (TEs) may be cis-regulatory elements to control gene expression. Here, mouse-specific TEs are shown as binding sites for multiple collaborating transcription factors in embryonic stem cells, and act as cis-regulatory modules in synergistic fashion.
- Published
- 2017
7. Selection of RARA-Positive Newly Diagnosed Unfit AML Patients with Elevated RARA Gene Expression Enriches for Features Associated with Primary Resistance to Venetoclax and Clinical Response to SY-1425, a Potent and Selective RARα Agonist, Plus Azacitidine
- Author
-
Li Zhou, Matthew L. Eaton, Kate Madigan, Qing Kang-Fortner, Graeme Hodgson, Michael Kelly, Angela Volkert, David A. Roth, and Christopher Fiore
- Subjects
Agonist ,Venetoclax ,medicine.drug_class ,business.industry ,Immunology ,Azacitidine ,Cell Biology ,Hematology ,Newly diagnosed ,Biochemistry ,chemistry.chemical_compound ,chemistry ,Gene expression ,Cancer research ,medicine ,business ,Selection (genetic algorithm) ,medicine.drug - Abstract
Introduction: Super-enhancer (SE) mapping in non-APL AML patient (pt) blasts identified RARα as a novel therapeutic target in approximately 30% of pts, who have elevated RARA gene expression. It was observed that the enhancer profile of this novel pt segment, where RARA expression was elevated, overlapped with the profile of mature monocytes (McKeown, 2017). Recently, several reports describe AML with monocytic features associated with resistance to venetoclax (Ven), a BCL-2 inhibitor that has emerged as a standard of care for treatment of pts with newly diagnosed (ND) unfit AML in combination with hypomethylating agents (HMAs) (Zhang, 2018; Kuusanmäki, 2019; Pei, 2020). Approximately one-third of pts do not respond to Ven plus HMAs including azacitidine (Aza) (DiNardo, 2019 and 2020), highlighting a continuing significant unmet need in ND unfit AML. SY-1425, a potent and selective RARα agonist, is in development for non-APL AML in combination with Aza and has demonstrated clinical activity with high rates of complete remission (CR) and deep CRs in RARA-positive (RARA+) ND unfit AML (DeBotton, 2019). Based on the overlap of monocytic features with RARA gene expression, we evaluated clinical samples of pts treated with SY-1425 plus Aza to correlate features of Ven resistance with the RARA biomarker and with clinical response to SY-1425 plus Aza. Methods: RARA gene expression in non-APL AML was evaluated in the TCGA and Beat AML RNA-seq datasets. AUC of cell viability curves were used to evaluate ex vivo sensitivity to compounds, including Ven, in the Beat AML dataset. A monocytic expression signature (MES) was developed using the expression of monocytic and primitive RNA markers in the TCGA dataset to analyze the monocytic phenotype. The MES used a logistic regression model with lasso regularization to distinguish FAB M4/5 (monocytic) from FAB M0/1/2 (primitive) using 10-fold cross-validation with 85% sensitivity and 80% specificity. The MES was then applied to the RNA-seq datasets from Beat AML and AML blasts from ND unfit AML pts in the ongoing SY-1425 plus Aza trial (NCT02807558), in which RARA+ pts were determined by an RT-qPCR based biomarker clinical trial assay (CTA). The MES, RARA expression, and Ven resistance-associated features were compared using Spearman's rho correlation; the association of the MES with the RARA biomarker and with IWG clinical responses in SY-1425 plus Aza treated pts was evaluated. Results: Analysis of RNA-seq in TCGA non-APL AML pts demonstrated higher RARA expression in monocytic AML (FAB M4/M5) than primitive AML (FAB M0/M1/M2) (p We further elucidated the relationships of RARA expression, AML monocytic phenotypes, and Ven resistance. Of 121 inhibitors tested ex vivo in primary Beat AML pt samples, Ven was the inhibitor most associated with treatment resistance in RARA+ vs. RARA- samples. Additionally, MES (rho=0.58), RARA (rho=0.48) and BCL-2 expression (rho=-0.49) had similar magnitude of association with ex vivo Ven resistance, with RARA+ samples showing much lower ex vivo sensitivity to Ven than RARA- samples (p=3×10-8). In 12 AML pt samples (Pei, 2020) treated with Ven ± Aza ex vivo, RARA expression was higher in the monocytic leukemia stem cells resistant to Ven ± Aza (p=0.005). To evaluate whether the RARA+ ND unfit AML pts in the ongoing SY-1425 plus Aza clinical trial were enriched for the monocytic phenotype of Ven resistance, RNA-seq was performed on enrolled pt AML blasts. Among 51 treated pts, 86% (19/22) of RARA+ and 83% (24/29) of RARA- pts yielded RNA-seq results. RARA+ pts were more monocytic than RARA- pts, as demonstrated by higher MES (p=0.002), with higher MCL-1 (p=0.001), and lower BCL-2, CD34, and CD117 expression (p=0.03, 8×10-6, 2×10-4, respectively). In pts with the best IWG response of CR/CRi, RARA+ pts (n=10) had higher MES than RARA- pts (n=8) (p=0.008). Conclusion: In ND unfit AML, RARA+ pts, including those with clinical responses to SY-1425 plus Aza, are enriched for monocytic features associated with resistance to Ven. SY-1425 plus Aza shows potential as a novel targeted regimen for the treatment of RARA+ ND unfit AML and warrants further development in this genomically defined subset of AML pts who may be resistant to upfront SOC therapy with Ven. Disclosures Fiore: Syros Pharmaceuticals, Inc.: Current Employment, Current equity holder in publicly-traded company. Kelly:Syros Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Volkert:Syros Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Zhou:Incyte: Current equity holder in publicly-traded company, Ended employment in the past 24 months; Syros Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Madigan:Syros Pharmaceuticals, Inc.: Current Employment, Current equity holder in publicly-traded company. Eaton:Syros Pharmaceuticals, Inc.: Current Employment, Current equity holder in publicly-traded company. Hodgson:Syros Pharmaceuticals, Inc.: Current Employment, Current equity holder in publicly-traded company. Roth:Syros Pharmaceuticals: Current Employment, Current equity holder in publicly-traded company. Kang-Fortner:Syros Pharmaceuticals, Inc.: Current Employment, Current equity holder in publicly-traded company.
- Published
- 2020
8. Intrinsic cell-penetrating activity propels Omomyc from proof of concept to viable anti-MYC therapy
- Author
-
Erika Serrano del Pozo, Christopher Fiore, Miguel Ángel Morcillo Alonso, Loïka Maltais, Sílvia Casacuberta-Serra, Marie-Eve Beaulieu, Danny Létourneau, Laura Soucek, Génesis Martín, Peter B. Rahl, Laia Foradada, Marta Oteo, Sandra Martínez-Martín, Eduardo Romero Sanz, Jonathan Whitfield, Virginia Castillo Cano, Toni Jauset, Matthew G. Guenther, Martin Montagne, Cynthia Tremblay, Meritxell Sánchez-Hervás, Daniel Massó-Vallés, Pierre Lavigne, and Mariano F. Zacarias-Fluck
- Subjects
Lung Neoplasms ,Transgene ,Cell ,Adenocarcinoma of Lung ,Cell-Penetrating Peptides ,E-Box Elements ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Promoter Regions, Genetic ,030304 developmental biology ,0303 health sciences ,business.industry ,Cancer ,DNA ,General Medicine ,medicine.disease ,Peptide Fragments ,In vitro ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,Basic-Leucine Zipper Transcription Factors ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Systemic administration ,Cancer research ,Female ,Protein Multimerization ,business ,Protein Binding - Abstract
Inhibiting MYC has long been considered unfeasible, although its key role in human cancers makes it a desirable target for therapeutic intervention. One reason for its perceived undruggability was the fear of catastrophic side effects in normal tissues. However, we previously designed a dominant-negative form of MYC called Omomyc and used its conditional transgenic expression to inhibit MYC function both in vitro and in vivo. MYC inhibition by Omomyc exerted a potent therapeutic impact in various mouse models of cancer, causing only mild, well-tolerated, and reversible side effects. Nevertheless, Omomyc has been so far considered only a proof of principle. In contrast with that preconceived notion, here, we show that the purified Omomyc mini-protein itself spontaneously penetrates into cancer cells and effectively interferes with MYC transcriptional activity therein. Efficacy of the Omomyc mini-protein in various experimental models of non-small cell lung cancer harboring different oncogenic mutation profiles establishes its therapeutic potential after both direct tissue delivery and systemic administration, providing evidence that the Omomyc mini-protein is an effective MYC inhibitor worthy of clinical development.
- Published
- 2019
- Full Text
- View/download PDF
9. Interactions between pluripotency factors specifycis-regulation in embryonic stem cells
- Author
-
Christopher Fiore and Barak A. Cohen
- Subjects
0301 basic medicine ,genetic processes ,Kruppel-Like Transcription Factors ,Gene Expression ,Regulatory Sequences, Nucleic Acid ,Biology ,Kruppel-Like Factor 4 ,Mice ,03 medical and health sciences ,Gene expression ,Genetics ,Animals ,natural sciences ,Genomic library ,Binding site ,Transcription factor ,Cells, Cultured ,Genetics (clinical) ,Gene Library ,Regulation of gene expression ,Binding Sites ,Models, Genetic ,Research ,fungi ,Mouse Embryonic Stem Cells ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,Regulatory sequence ,KLF4 ,Thermodynamics ,Transcription Factors - Abstract
We investigated how interactions between pluripotency transcription factors (TFs) affectcis-regulation. We created hundreds of syntheticcis-regulatory elements (CREs) comprised of combinations of binding sites for pluripotency TFs and measured their expression in mouse embryonic stem (ES) cells. A thermodynamic model that incorporates interactions between TFs explains a large portion (72%) of the variance in expression of these CREs. These interactions include three favorable heterotypic interactions between TFs. The model also predicts an unfavorable homotypic interaction between TFs, helping to explain the observation that homotypic chains of binding sites express at low levels. We further investigated the expression driven by CREs comprised of homotypic chains of KLF4 binding sites. Our results suggest that KLF homologs make unique contributions to regulation by these CREs. We conclude that a specific set of interactions between pluripotency TFs plays a large role in setting the levels of expression driven by CREs in ES cells.
- Published
- 2016
10. Abstract P4-04-02: Epigenomic analysis of cancer stem cells (CSCs) from triple-negative breast cancer (TNBC) reveals p63 and p73 as core metastasis drivers
- Author
-
Elisha Verhaar, Brian Bierie, Robert A. Weinberg, Matthew G. Guenther, Daneyal Farouq, Yogesh Chutake, Bingbing Yuan, Eric N. Olson, Ian Engstrom, Ferenc Reinhardt, John Carulli, Arthur W. Lambert, Mei Wei Chen, Joana Liu Donaher, and Christopher Fiore
- Subjects
Cancer Research ,Cancer ,Epigenome ,Biology ,medicine.disease ,Metastasis ,Transcriptome ,Breast cancer ,Oncology ,Cancer stem cell ,Cancer research ,medicine ,Triple-negative breast cancer ,Epigenomics - Abstract
Cancer stem cells (CSCs) are key drivers of cancer metastasis, drug resistance, and disease recurrence. While the transcriptional regulatory circuitry that underlies the generation and propagation of the breast CSC state is not completely understood, defining the core regulators of the CSC state has the potential to reveal how these cells arise and point to potential targetable pathways for therapeutic intervention. We isolated CSCs by selecting for surface expression of integrin β4 (ITGB4). ITGB4-hi cells are highly enriched for CSCs and display cellular and molecular features indicative of their residence in an intermediate EMT state, including the ability to seed metastasis in vivo. Using ITGB4-hi CSCs isolated from the SUM159 breast cancer cell line, we deployed epigenome mapping by H3K27ac ChIP-seq, nucleosome occupancy by ATAC-seq, and transcriptome analysis by RNA-seq. Using comparative analysis of enhancer usage and DNA accessibility, we identified the transcriptional regulatory circuitry of breast cancer CSCs and pinpointed the master transcription factors that control the CSC state. We find that p63/p73 proteins function as master transcriptional regulators in breast CSCs and establish super-enhancers at critical CSC state genes. Loss of p63 and p73 disrupts the transcriptional circuitry of CSCs and leads to a highly mesenchymal state that is incompatible with metastatic colonization. Re-introduction of p63/p73 expression into non-CSCs leads to reactivation of the CSC transcriptional program and enables metastasis. These findings reveal the unique transcriptional circuitry of CSCs and suggest potential dependencies and pathways for therapeutic targeting of cells in the CSC state. Citation Format: Arthur Lambert, Christopher Fiore, Yogesh Chutake, Elisha Verhaar, Mei Wei Chen, Joana Donaher, Ferenc Reinhardt, Ian Engstrom, Bingbing Yuan, Brian Bierie, Daneyal Farouq, John Carulli, Eric Olson, Matthew Guenther, Robert Weinberg. Epigenomic analysis of cancer stem cells (CSCs) from triple-negative breast cancer (TNBC) reveals p63 and p73 as core metastasis drivers [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P4-04-02.
- Published
- 2020
11. A Tale of Two Anomalies: Higher Returns of Low-Risk Stocks and Return Seasonality
- Author
-
Christopher Fiore and Atanu Saha
- Subjects
Economics and Econometrics ,Low-volatility anomaly ,Financial economics ,Risk–return spectrum ,Sell in May ,Treasury ,Economics ,Econometrics ,Portfolio ,Capital asset pricing model ,Volatility (finance) ,health care economics and organizations ,Finance ,Stock (geology) - Abstract
Prior studies have shown that low beta and low volatility stocks earn higher average returns than high beta and high volatility stocks, contradicting the prediction of the capital asset pricing model and the fundamental relationship between risk and return. In this paper, we demonstrate that this phenomenon is driven by the seasonality of stock returns. We show that the risk-return tradeoff does hold in the nonsummer months, and that switching to a portfolio of low-risk stocks in summer outperforms—both in terms of absolute and in risk-adjusted returns—buy and hold strategies as well as the Sell in May strategy of switching to treasury bills in summer.
- Published
- 2015
12. High-throughput functional testing of ENCODE segmentation predictions
- Author
-
Christopher Fiore, Jamie C. Kwasnieski, Hemangi G. Chaudhari, and Barak A. Cohen
- Subjects
Regulation of gene expression ,Genetics ,Models, Genetic ,biology ,Sequence Analysis, RNA ,Research ,Computational Biology ,Enhancer RNAs ,Regulatory Sequences, Nucleic Acid ,ENCODE ,Histones ,Logistic Models ,Histone ,Gene Expression Regulation ,biology.protein ,Humans ,K562 Cells ,Enhancer ,Gene ,Transcription factor ,Embryonic Stem Cells ,Genetics (clinical) ,Human embryonic stem cell line - Abstract
The histone modification state of genomic regions is hypothesized to reflect the regulatory activity of the underlying genomic DNA. Based on this hypothesis, the ENCODE Project Consortium measured the status of multiple histone modifications across the genome in several cell types and used these data to segment the genome into regions with different predicted regulatory activities. We measured the cis-regulatory activity of more than 2000 of these predictions in the K562 leukemia cell line. We tested genomic segments predicted to be Enhancers, Weak Enhancers, or Repressed elements in K562 cells, along with other sequences predicted to be Enhancers specific to the H1 human embryonic stem cell line (H1-hESC). Both Enhancer and Weak Enhancer sequences in K562 cells were more active than negative controls, although surprisingly, Weak Enhancer segmentations drove expression higher than did Enhancer segmentations. Lower levels of the covalent histone modifications H3K36me3 and H3K27ac, thought to mark active enhancers and transcribed gene bodies, associate with higher expression and partly explain the higher activity of Weak Enhancers over Enhancer predictions. While DNase I hypersensitivity (HS) is a good predictor of active sequences in our assay, transcription factor (TF) binding models need to be included in order to accurately identify highly expressed sequences. Overall, our results show that a significant fraction (∼26%) of the ENCODE enhancer predictions have regulatory activity, suggesting that histone modification states can reflect the cis-regulatory activity of sequences in the genome, but that specific sequence preferences, such as TF-binding sites, are the causal determinants of cis-regulatory activity.
- Published
- 2014
13. Model-based transcriptome engineering promotes a fermentative transcriptional state in yeast
- Author
-
Michael R. Brent, Christopher Fiore, Randall H. Brown, Holly Brown, Stacey R. Gish, Ezekiel J. Maier, and Drew G. Michael
- Subjects
0301 basic medicine ,Saccharomyces cerevisiae Proteins ,Saccharomyces cerevisiae ,Biology ,Xylose ,Metabolic engineering ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Transcriptional regulation ,Gene Regulatory Networks ,Multidisciplinary ,Ethanol ,Models, Genetic ,biology.organism_classification ,Yeast ,030104 developmental biology ,Glucose ,Biochemistry ,chemistry ,PNAS Plus ,Metabolic Engineering ,Fermentation ,Flux (metabolism) ,030217 neurology & neurosurgery ,Algorithms ,Gene Deletion ,Transcription Factors - Abstract
The ability to rationally manipulate the transcriptional states of cells would be of great use in medicine and bioengineering. We have developed an algorithm, NetSurgeon, which uses genome-wide gene-regulatory networks to identify interventions that force a cell toward a desired expression state. We first validated NetSurgeon extensively on existing datasets. Next, we used NetSurgeon to select transcription factor deletions aimed at improving ethanol production in Saccharomyces cerevisiae cultures that are catabolizing xylose. We reasoned that interventions that move the transcriptional state of cells using xylose toward that of cells producing large amounts of ethanol from glucose might improve xylose fermentation. Some of the interventions selected by NetSurgeon successfully promoted a fermentative transcriptional state in the absence of glucose, resulting in strains with a 2.7-fold increase in xylose import rates, a 4-fold improvement in xylose integration into central carbon metabolism, or a 1.3-fold increase in ethanol production rate. We conclude by presenting an integrated model of transcriptional regulation and metabolic flux that will enable future efforts aimed at improving xylose fermentation to prioritize functional regulators of central carbon metabolism.
- Published
- 2016
14. Vitamin D Receptor Protein Expression in Tumor Tissue and Prostate Cancer Progression
- Author
-
Richard Flavin, Michelangelo Fiorentino, Kathryn L. Penney, Christopher Fiore, Whitney K. Hendrickson, Rosina T. Lis, Lorelei A. Mucci, Julie L. Kasperzyk, Massimo Loda, Meir J. Stampfer, Edward Giovannucci, Fang Fang, Philip W. Kantoff, Jing Ma, Hendrickson WK, Flavin R, Kasperzyk JL, Fiorentino M, Fang F, Lis R, Fiore C, Penney KL, Ma J, Kantoff PW, Stampfer MJ, Loda M, Mucci LA, and Giovannucci E
- Subjects
Male ,gene polymorphisms ,PCA3 ,Cancer Research ,medicine.medical_specialty ,Oncogene Proteins, Fusion ,d-3 ,united-states ,Single-nucleotide polymorphism ,urologic and male genital diseases ,Calcitriol receptor ,Prostate cancer ,1,25-dihydroxyvitamin-d ,subsequent development ,Internal medicine ,Original Reports ,medicine ,Vitamin D and neurology ,Humans ,Prospective Studies ,Vitamin D ,physicians health ,Prospective cohort study ,Aged ,risk ,biology ,business.industry ,Incidence ,Prostatic Neoplasms ,Prostate-Specific Antigen ,medicine.disease ,cytosolic calcium ,FokI ,d metabolites ,Prostate-specific antigen ,prostate cancer vitamin d gleason grade ,Endocrinology ,Oncology ,Disease Progression ,biology.protein ,Cancer research ,Receptors, Calcitriol ,kinase-c ,business - Abstract
Purpose Data suggest that circulating 25-hydroxyvitamin D [25(OH)D] interacts with the vitamin D receptor (VDR) to decrease proliferation and increase apoptosis for some malignancies, although evidence for prostate cancer is less clear. How VDR expression in tumor tissue may influence prostate cancer progression has not been evaluated in large studies. Patients and Methods We examined protein expression of VDR in tumor tissue among 841 patients with prostate cancer in relation to risk of lethal prostate cancer within two prospective cohorts, the Physicians' Health Study and Health Professionals Follow-Up Study. We also examined the association of VDR expression with prediagnostic circulating 25(OH)D and 1,25-dihydroxyvitamin D levels and with two VDR single nucleotide polymorphisms, FokI and BsmI. Results Men whose tumors had high VDR expression had significantly lower prostate-specific antigen (PSA) at diagnosis (P for trend < .001), lower Gleason score (P for trend < .001), and less advanced tumor stage (P for trend < .001) and were more likely to have tumors harboring the TMPRSS2:ERG fusion (P for trend = .009). Compared with the lowest quartile, men whose tumors had the highest VDR expression had significantly reduced risk of lethal prostate cancer (hazard ratio [HR], 0.17; 95% CI, 0.07 to 0.41). This association was only slightly attenuated after adjustment for Gleason score and PSA at diagnosis (HR, 0.33; 95% CI, 0.13 to 0.83) or, additionally, for tumor stage (HR, 0.37; 95% CI, 0.14 to 0.94). Neither prediagnostic plasma vitamin D levels nor VDR polymorphisms were associated with VDR expression. Conclusion High VDR expression in prostate tumors is associated with a reduced risk of lethal cancer, suggesting a role of the vitamin D pathway in prostate cancer progression.
- Published
- 2011
15. Fatty Acid Synthase Polymorphisms, Tumor Expression, Body Mass Index, Prostate Cancer Risk, and Survival
- Author
-
Stephen P. Finn, Meir J. Stampfer, Giuseppe Fedele, Fredrick R. Schumacher, Christopher Fiore, Anna Eisenstein, Weiliang Qiu, Edward Giovannucci, Jing Ma, Kathryn L. Penney, Michelangelo Fiorentino, Paul L. Nguyen, Lorelei A. Mucci, Rosina T. Lis, Matthew L. Freedman, Massimo Loda, Jorge E. Chavarro, Nguyen PL, Ma J, Chavarro JE, Freedman ML, Lis R, Fedele G, Fiore C, Qiu W, Fiorentino M, Finn S, Penney KL, Eisenstein A, Schumacher FR, Mucci LA, Stampfer MJ, Giovannucci E, and Loda M
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Single-nucleotide polymorphism ,Overweight ,urologic and male genital diseases ,Polymorphism, Single Nucleotide ,Body Mass Index ,Prostate cancer ,Risk Factors ,Internal medicine ,Original Reports ,medicine ,Humans ,SNP ,Genetic Predisposition to Disease ,Aged ,biology ,business.industry ,Prostatic Neoplasms ,Cancer ,Middle Aged ,medicine.disease ,Fatty acid synthase ,Endocrinology ,Oncology ,Relative risk ,biology.protein ,fatty acid synthase body mass index prostate cancer ,Fatty Acid Synthases ,medicine.symptom ,business ,Body mass index - Abstract
Purpose Fatty acid synthase (FASN) regulates de novo lipogenesis, body weight, and tumor growth. We examined whether common germline single nucleotide polymorphisms (SNPs) in the FASN gene affect prostate cancer (PCa) risk or PCa-specific mortality and whether these effects vary by body mass index (BMI). Methods In a prospective nested case-control study of 1,331 white patients with PCa and 1,267 age-matched controls, we examined associations of five common SNPs within FASN (and 5 kb upstream/downstream, R2 > 0.8) with PCa incidence and, among patients, PCa-specific death and tested for an interaction with BMI. Survival analyses were repeated for tumor FASN expression (n = 909). Results Four of the five SNPs were associated with lethal PCa. SNP rs1127678 was significantly related to higher BMI and interacted with BMI for both PCa risk (Pinteraction = .004) and PCa mortality (Pinteraction = .056). Among overweight men (BMI ≥ 25 kg/m2), but not leaner men, the homozygous variant allele carried a relative risk of advanced PCa of 2.49 (95% CI, 1.00 to 6.23) compared with lean men with the wild type. Overweight patients carrying the variant allele had a 2.04 (95% CI, 1.31 to 3.17) times higher risk of PCa mortality. Similarly, overweight patients with elevated tumor FASN expression had a 2.73 (95% CI, 1.05 to 7.08) times higher risk of lethal PCa (Pinteraction = .02). Conclusion FASN germline polymorphisms were significantly associated with risk of lethal PCa. Significant interactions of BMI with FASN polymorphisms and FASN tumor expression suggest FASN as a potential link between obesity and poor PCa outcome and raise the possibility that FASN inhibition could reduce PCa-specific mortality, particularly in overweight men.
- Published
- 2010
16. Abstract PR05: SY-1425 (tamibarotene), a potent and selective RARα agonist, induces changes in the transcriptional regulatory circuit of AML cells leading to differentiation
- Author
-
Christian C. Fritz, Matthew L. Eaton, Mei Wei Chen, Eric N. Olson, Matthew G. Guenther, M. Ryan Corces, Emily Lee, Darren Smith, Ravindra Majeti, Kathryn Austgen, Christopher Fiore, and Michael R. McKeown
- Subjects
Cancer Research ,Myeloid ,Biology ,chemistry.chemical_compound ,Haematopoiesis ,medicine.anatomical_structure ,Oncology ,RUNX1 ,chemistry ,Retinoic acid receptor alpha ,medicine ,Cancer research ,IRF8 ,Progenitor cell ,Enhancer ,Transcription factor - Abstract
SY-1425 (tamibarotene) is a potent and selective agonist of the transcription factor (TF) retinoic acid receptor alpha (RARα) that is currently being evaluated in a biomarker-directed Ph2 clinical study in AML and MDS. A subset of AML and MDS patients, referred to as RARA-high, has previously been found to have a large enhancer (super-enhancer) at the RARA locus or upregulation of IRF8, a RARα associated TF. Here, we profile the noncoding genome and transcriptional landscape in AML cells to define the circuitry of RARA-high AML characterized by RARα pathway activation. In addition, AML cell lines were profiled with and without SY-1425 treatment to query the perturbations of key regulatory connections by SY-1425. To better understand the regulatory circuitry of the RARα pathway in AML, we profiled RARA-high and RARA-low AML cell lines with a combination of ATAC-seq, H3K27ac ChIP-seq, and transcriptomic profiling. Using the enhancer landscape of these cell lines compared to a set of healthy human immune cells comprising hematopoietic stem and progenitor cells (HSPCs) as well as multiple myeloid lineages, we performed a non-negative least squares (NNLS) regression to determine the hematopoietic lineage components of these AML cell lines. We found that RARA-high cell lines sensitive to SY-1425 (THP-1, OCI-AML3, MV4-11, and EOL-1) tend to be most similar to monocytes, while RARA-low cell lines insensitive to SY-1425 (KG-1a, Kasumi-1, and OCI-M1) are more HSPC or erythroid-like. We then studied the effect of SY-1425 on the differentiation state of these AML cell lines by additionally profiling them with and without treatment. We find that the enhancer landscape of RARA-high AML cell lines was changed by SY-1425 toward one resembling more fully differentiated myeloid cells of different subtypes, including dendritic cells, granulocytic cells, and macrophages. This was in contrast to the well-characterized granulocytic differentiation of APL caused by retinoids. Functional validation by flow cytometry confirmed surface marker changes consistent with the observed epigenomic alterations. Comparable changes were not found in RARA-low AML cell lines. This effect supports the hypothesis that SY-1425 treatment relieves a RARα-mediated differentiation block of RARA-high AML. To understand the individual regulatory connections being modulated in response to SY-1425, we investigated the binding locations of TFs in the RARA-high cell lines with and without SY-1425 treatment. As expected, enhancers and genes bound by RARα were associated with upregulation of transcription in response to SY-1425. TF binding sites for IRF1 and IRF8 were also identified at induced enhancers. Conversely, known factors important for maintaining an immature/proliferative state were found associated with enhancers most deactivated by SY-1425 (as measured by H3K27ac loss). These included RUNX1, CEBP, and members of the FOS/JUN circuit (which were noted previously as a component of the oncogenic RARα circuit in patient samples). Taken together, these results support that the mechanism of action for SY-1425 is through perturbation of myeloid regulatory relationships in RARA-high AML blasts, leading to a more differentiated phenotype. Based on the understanding of AML- and RARα-driven tumor circuitry, we have generated insight into morphologic, lineage marker, and target-gene based measures to be used in conjunction with clinical studies. The ongoing Ph2 study of SY-1425 in AML and MDS (NCT02807558) will explore measurement of these features in patients. This abstract is also being presented as Poster 06. Citation Format: Christopher M. Fiore, Michael R. McKeown, Emily Lee, Matthew L. Eaton, Darren Smith, Kathryn Austgen, Mei Wei Chen, Matthew Guenther, M. Ryan Corces, Ravindra Majeti, Eric Olson, Christian C. Fritz. SY-1425 (tamibarotene), a potent and selective RARα agonist, induces changes in the transcriptional regulatory circuit of AML cells leading to differentiation [abstract]. In: Proceedings of the Second AACR Conference on Hematologic Malignancies: Translating Discoveries to Novel Therapies; May 6-9, 2017; Boston, MA. Philadelphia (PA): AACR; Clin Cancer Res 2017;23(24_Suppl):Abstract nr PR05.
- Published
- 2017
17. c-Jun amplification and overexpression are oncogenic in liposarcoma but not always sufficient to inhibit the adipocytic differentiation programme
- Author
-
Eric L. Snyder, Jonathan A. Fletcher, Deborah J. Sandstrom, Ewa Sicinska, Kenneth Law, Paola Dal Cin, Massimo Loda, Scott J. Rodig, Christopher Fiore, Joseph Brito, Christopher D.M. Fletcher, and Dyane Bailey
- Subjects
medicine.diagnostic_test ,Kinase ,Cellular differentiation ,c-jun ,Biology ,Liposarcoma ,medicine.disease_cause ,medicine.disease ,Pathology and Forensic Medicine ,body regions ,Small hairpin RNA ,Cancer research ,medicine ,biology.protein ,Mdm2 ,Carcinogenesis ,neoplasms ,Fluorescence in situ hybridization - Abstract
Genomic amplification of c-Jun and its upstream kinases have been implicated as a mechanism of progression from well-differentiated to dedifferentiated liposarcoma. To further define the role of c-Jun in liposarcoma progression, we performed immunohistochemistry for c-Jun and its activating kinase ASK1 on a series of liposarcomas (n = 81). We correlated the results with fluorescence in situ hybridization to detect c-Jun amplification. We also derived new cell lines from dedifferentiated liposarcomas with c-Jun amplification. c-Jun protein is expressed in the majority of dedifferentiated liposarcomas (91%) and their well-differentiated components (59%), but only in the minority of pure well-differentiated liposarcomas (27%). When c-Jun is amplified in dedifferentiated liposarcoma, it is interspersed with amplified MDM2 on ring and giant marker chromosomes. MDM2 amplification is one of the earliest events in liposarcoma development, and these results suggest that c-Jun was amplified at a similar time in the evolution of the tumour. In addition, shRNA to c-Jun in c-Jun-amplified liposarcoma cells reduces cell number in vitro and inhibits tumour formation in vivo without an observable effect on the differentiation state of the liposarcoma cells. Thus, c-Jun amplification is oncogenic in liposarcomas but not always sufficient for inhibition of adipocytic differentiation.
- Published
- 2009
18. Examining Success
- Author
-
Jonathan C. Lipson and Christopher Fiore Marotta
- Published
- 2015
19. Abstract 2167: Preclinical validation of an Omomyc cell-penetrating peptide as a viable anti-Myc therapy
- Author
-
Christopher Fiore, Peter B. Rahl, Marta Oteo Vives, Matthew G. Guenther, Daniel Massó-Vallés, Miguel Ángel Morcillo Alonso, Martin Montagne, Sandra Martínez-Martín, Cynthia Tremblay, Pierre Lavigne, Jonathan Whitfield, Mariano F. Zacarias-Fluck, Marie-Eve Beaulieu, Laura Soucek, Loïka Maltais, Laia Foradada, Erika Serrano del Pozo, Toni Jauset, Silvia Casacuberta, and Eduardo Romero Sanz
- Subjects
Cancer Research ,Tumor microenvironment ,Cell growth ,Genetic enhancement ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Transactivation ,Oncology ,Cancer cell ,Cancer research ,medicine ,Transcriptional regulation ,Carcinogenesis - Abstract
Deregulation of the MYC oncoprotein promotes tumorigenesis in most, if not all, cancers and is often associated with poor prognosis. However, targeting MYC has long been considered impossible based on the assumption that it would cause catastrophic side effects in normal tissues. Despite this general preconceived notion, we showed that MYC inhibition exerts extraordinary therapeutic impact in various genetic mouse models of cancer, and causes only mild, well-tolerated and reversible side effects. For these studies we employed the systemic and conditional expression of a dominant negative of MYC, called Omomyc, which we designed and validated, and that can inhibit MYC transactivation function both in vitro and in vivo. To date, Omomyc has only been considered a proof of principle, with any potential clinical application limited to gene therapy. Here we actually show that the 11 kDa Omomyc polypeptide spontaneously transduces into cancer cells, demonstrating unexpected cell-penetrating ability. Once inside the nuclei, the polypeptide effectively blocks MYC binding to its target DNA sites, interfering with MYC transcriptional regulation and halting cell proliferation. Moreover, intranasal (i.n.) administration of the Omomyc polypeptide in mice results in its rapid and persistent distribution to lungs, as well as to other organs (i.e. intestine, liver, kidneys and brain). Importantly, i.n. treatment of mice bearing either Non-Small-Cell-Lung-Cancer (NSCLC) or glioblastoma (GBM) with the Omomyc cell-penetrating peptide (OmomycCPP) significantly reduces tumor burden compared to their control counterparts. Notably, tumor regression is accompanied by significant reprogramming of the tumor microenvironment and tumor immune response. In summary, our data indicate that this novel generation of polypeptides represents a new opportunity to potentially inhibit MYC pharmacologically in a variety of malignant diseases. Citation Format: Marie-eve Beaulieu, Toni Jauset, Daniel Massó-Vallés, Peter Rahl, Sandra Martinez-Martin, Loika Maltais, Mariano F. Zacarias-Fluck, Silvia Casacuberta, Erika Serrano del Pozo, Christopher Fiore, Laia Foradada, Matthew Guenther, Eduardo Romero Sanz, Marta Oteo Vives, Cynthia Tremblay, Martin Montagne, Miguel Ángel Morcillo Alonso, Jonathan R. Whitfield, Pierre Lavigne, Laura Soucek. Preclinical validation of an Omomyc cell-penetrating peptide as a viable anti-Myc therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2167. doi:10.1158/1538-7445.AM2017-2167
- Published
- 2017
20. SPINK1 protein expression and prostate cancer progression
- Author
-
Lorelei A. Mucci, Whitney K. Hendrickson, Gregory L Judson, Edward C. Stack, Christopher S. Sweeney, Dyane Bailey, Andreas Pettersson, Meir J. Stampfer, Elizabeth Nuttall, Stephen P. Finn, Edward Giovannucci, Philip W. Kantoff, Neil E. Martin, Katja Fall, Rosina T. Lis, Michelangelo Fiorentino, Howard D. Sesso, Jennifer A. Sinnott, Lauren M. Kunz, Massimo Loda, Jennifer R. Rider, Richard Flavin, Christopher Fiore, Kathryn L. Penney, Flavin RJ, Pettersson A, Hendrickson WK, Fiorentino M, Finn SP, Kunz L, Judson G, Lis RT, Bailey D, Fiore C, Nuttall EJ, Martin NE, Stack EC, Penney KL, Rider JR, Sinnott JA, Sweeney CS, Sesso HD, Fall K, Giovannucci EL, Kantoff PW, Stampfer MJ, Loda M, and Mucci LA
- Subjects
Oncology ,PCA3 ,Biochemical recurrence ,Male ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Adenocarcinoma ,TMPRSS2 ,Article ,Prosdtate cancer, SPINK1 ,Prostate cancer ,Prostate ,Internal medicine ,Biomarkers, Tumor ,Medicine ,PTEN ,Humans ,Aged ,Proportional Hazards Models ,Prostatectomy ,biology ,business.industry ,Cancer ,Prostatic Neoplasms ,Middle Aged ,medicine.disease ,Immunohistochemistry ,3. Good health ,medicine.anatomical_structure ,Tissue Array Analysis ,Trypsin Inhibitor, Kazal Pancreatic ,biology.protein ,Disease Progression ,Neoplasm Recurrence, Local ,business ,Carrier Proteins - Abstract
Purpose: SPINK1 overexpression has been described in prostate cancer and is linked with poor prognosis in many cancers. The objective of this study was to characterize the association between SPINK1 overexpression and prostate cancer–specific survival. Experimental Design: The study included 879 participants in the U.S. Physicians' Health Study and Health Professionals Follow-Up Study, diagnosed with prostate cancer (1983–2004) and treated by radical prostatectomy. Protein tumor expression of SPINK1 was evaluated by immunohistochemistry on tumor tissue microarrays. Results: Seventy-four of 879 (8%) prostate cancer tumors were SPINK1 positive. Immunohistochemical data were available for PTEN, p-Akt, pS6, stathmin, androgen receptor (AR), and ERG (as a measure of the TMPRSS2:ERG translocation). Compared with SPINK1-negative tumors, SPINK1-positive tumors showed higher PTEN and stathmin expression, and lower expression of AR (P < 0.01). SPINK1 overexpression was seen in 47 of 427 (11%) ERG-negative samples and in 19 of 427 (4%) ERG-positive cases (P = 0.0003). We found no significant associations between SPINK1 status and Gleason grade or tumor stage. There was no association between SPINK1 expression and biochemical recurrence (P = 0.56). Moreover, there was no association between SPINK1 expression and prostate cancer mortality (there were 75 lethal cases of prostate cancer during a mean of 13.5 years follow-up; HR = 0.71; 95% confidence interval, 0.29–1.76). Conclusions: Our results suggest that SPINK1 protein expression may not be a predictor of recurrence or lethal prostate cancer amongst men treated by radical prostatectomy. SPINK1 and ERG protein expression do not seem to be entirely mutually exclusive, as some previous studies have suggested. Clin Cancer Res; 20(18); 4904–11. ©2014 AACR.
- Published
- 2014
21. Clinical Pharmacodynamic Markers and Combinations with SY1425 (tamibarotene) in a Genomically-Defined Subset of Non-APL AML
- Author
-
Christian C. Fritz, Michael R. McKeown, Christopher Fiore, Matthew L. Eaton, and Emily Lee
- Subjects
Myeloid ,Cellular differentiation ,Immunology ,Azacitidine ,Cell ,Decitabine ,Cell Biology ,Hematology ,Pharmacology ,Biology ,Biochemistry ,chemistry.chemical_compound ,Retinoic acid receptor ,medicine.anatomical_structure ,chemistry ,medicine ,Cancer research ,Tamibarotene ,Transcription factor ,medicine.drug - Abstract
SY-1425, a potent and selective agonist of the retinoic acid receptor RARα, is being investigated in a Ph2 trial in a novel genomically-defined subset of non-APL AML and MDS patients (clinicaltrials.gov NCT02807558). RARa is a nuclear hormone receptor and transcription factor that regulates genes involved in cell differentiation and proliferation. We identified a super-enhancer (SE) at the RARA locus, the gene encoding RARa, in a subset of primary non-APL AML blasts. Preclinical models demonstrated a correlation between the presence of a RARA SE and sensitivity to SY-1425, providing the rationale for clinical investigation. Further research has investigated pharmacodynamics (PD) markers and combinations of drugs to support clinical development of SY-1425. In this study we identified DHRS3mRNA induction as a measure of RARα target engagement with SY-1425. We also demonstrated synergy in preclinical models with SY-1425 and hypomethylating agents. Since RARα is a transcription factor that regulates target genes when bound by a retinoid, we characterized the dynamic expression changes of a panel of RARA enhancer- high and - low non-APL AML cell lines (hereafter referred to as RARA-high and -low) in response to SY-1425 treatment. DHRS3 showed the largest expression increase following treatment in 3 RARA-high cell lines, with a range of 29 to 115 fold. In contrast, there was a much lower DHRS3 induction in 3 RARA-low cell lines (range of 1.6 to 6.1 fold). Induction was found to be both time- and dose-dependent with maximal induction at approximately 6 hours and half maximal induction near the EC50 for the anti-proliferative effect in RARA-high cell lines. DHRS3 encodes dehydrogenase/reductase (SDR family) member 3, a metabolic enzyme involved in maintaining cellular retinol homeostasis and had previously been shown to be induced by retinoids. Thus, DHRS3induction in tumor cells represents a potentially useful PD marker for clinical studies of SY-1425. To better understand the mechanism of induction of DHRS3 by SY-1425 we examined the chromosomal localization of RARα as well as the epigenomic state of the DHRS3 locus by ChIP-seq for RARα and H3K27 acetylation, the latter being an indicator of active enhancers and promoters. In the untreated state, OCI-AML3 (a typical RARA-high AML cell line) was found to have multiple RARα binding sites both within and distal to the DHRS3 gene but minimal H3K27 acetylation. Following treatment with SY-1425, the level of H3K27 acetylation at DHRS3 increased, resulting in the formation of a SE. Moreover, the SE encompassed the RARα binding sites, consistent with the model in which SY-1425 converts RARα into an activator of DHRS3expression. Similar results were seen for the CD38 locus in which SY-1425 treatment increased expression, H3K27 acetylation, and RARα binding. CD38 is a cell surface antigen and marker of myeloid maturation readily analyzed by FACS analysis, suggesting it could be an additional PD marker to be used in clinical studies. Indeed, it was found that SY-1425 induced CD38 cell surface expression at similar levels in RARA-high AML cell lines and the NB-4 APL cell line, but not in RARA-low cell lines. We also investigated combinations of SY-1425 with approved or investigational AML and MDS agents in in vitro and in vivo models to inform future clinical studies and to further explore potential PD markers unique to the combined action of the drugs. Several standard of care agents and drugs in current development were found to have synergistic interactions with SY-1425 in RARA-high but not RARA-low cell lines. In particular, azacitidine and decitabine each showed strong in vitro synergy with SY-1425. Evaluation of SY-1425 plus azacitidine in a RARA-high PDX model of non-APL AML demonstrated a better response compared to either agent alone. Additional genome-wide ChIP-seq and expression studies of RARA-high cells treated with various combinations are being investigated to identify optimal PD markers for these combinations. These studies support the use of DHRS3 mRNA induction in tumor cells as a PD marker in the recently initiated Ph2 study of SY-1425 in genomically-defined non-APL AML and MDS patients (clinicaltrials.gov NCT02807558) and further exploration as a PD marker for future combination studies. Disclosures McKeown: Syros Pharmaceuticals: Employment, Equity Ownership. Fiore:Syros Pharmaceuticals: Employment, Equity Ownership. Lee:Syros Pharmaceuticals: Employment, Equity Ownership. Eaton:Syros Pharmaceuticals: Employment, Equity Ownership. Fritz:Syros Pharmaceuticals: Employment, Equity Ownership.
- Published
- 2016
22. SY1425 (tamibarotene) Induces Profound Transcriptional Changes in AML Tumors with High Retinoic Acid Receptor Alpha
- Author
-
Emily Lee, Christian C. Fritz, Matthew L. Eaton, Michael R. McKeown, and Christopher Fiore
- Subjects
Genetics ,Gene knockdown ,Immunology ,Retinoic acid ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Chromatin ,Gene expression profiling ,chemistry.chemical_compound ,chemistry ,Retinoic acid receptor alpha ,Gene expression ,Cancer research ,Enhancer ,Transcription factor - Abstract
Retinoic acid receptor alpha (RARα) regulates myeloid differentiation and proliferation through the regulation of specific sets of genes. When unbound by a ligand, RARα is a repressive transcription factor while in its ligand-bound state it functions as a transcriptional activator. Previously, blast cells from a subset of individuals with non-APL AML were found to have a super-enhancer (SE), as revealed by H3K27 acetyl ChIP-Seq, associated with the RARA locus (hereafter called RARA-high), suggesting that tumor cell proliferation may have a dependency on RARA that can be exploited for therapeutic benefit. SEs are exceptionally large, highly active chromatin regions that are densely occupied by transcription factors and have been implicated in oncogene expression. Indeed, RARA-high non-APL AML cell lines showed >1000-fold increased sensitivity compared to RARA-low cells to the potent and selective RARα agonist SY-1425 (tamibarotene) as well as efficacy in non-APL AML patient derived xenograft models with a dependency on RARA. Since RARα is a transcription factor and the direct target of SY-1425, we investigated the change SY-1425 had on the transcriptional program of non-APL AML cell lines and the mechanism underlying those changes. Expression profiling on a panel of AML cell lines revealed that RARA-high AML cell lines had profound transcriptional changes in response to SY-1425, with 437 genes significantly changed, while RARA-low cell lines did not show significant gene expression changes. Gene set enrichment analysis (GSEA) of three RARA-high AML cell lines revealed that the genes upregulated by SY-1425 in the RARA-high cells are associated with immune signaling, interferon induction, protein secretion, and pathways associated with complement, MHC and integrin functions, all pathways indicative of more differentiated blood cells. Signatures downregulated by SY-1425 include MYC target genes. These findings are consistent with SY-1425 increasing the expression of genes involved in differentiation and decreasing those involved in proliferation. Genome-wide ChIP-Seq analysis revealed an increase in H3K27 acetylation at loci found to have strong RARα peaks as well as increased expression of those genes upon treatment with SY-1425. Together, these data support a model in which RARα binding nucleates functional enhancers in response to SY-1425 thereby upregulating proximal target gene expression and promotion of differentiation. The gene expression and epigenomic responses of RARA-high AML cell lines to SY-1425 were found to be similar to the responses of an APL cell line (NB-4) to retinoids or SY-1425. Gene sets identified in response to either retinoid treatment or genetic perturbation, such as forced expression or RAR-fusions or knockdown, matched the gene sets identified in RARA-high AML cell lines. Furthermore, the quantitative response of both NB-4 and RARA-high AML cell lines to SY-1425 was found to be similar. Across the genome, RARα binding was highly conserved between NB-4 and RARA-high AML cell lines with less overlap with the RARA-low cell lines. For example, the transcriptional and H3K27 acetylation alterations at the known PML-RARα target gene TGM2 following retinoid treatment was similar in NB-4 and the RARA- high cell lines. This locus also had a strong RARα binding site that is conserved among the cell lines and co-localized with a strong H3K27 acetylation peak. Consistent with the pattern of occupancy of RARα on the genome, the transcriptional response of the RARA enhancer-high cell lines to SY-1425 treatment was similar to the response of APL ex-vivo patient samples to retinoic acid treatment. These data support a model of a common biological response to retinoids between cells with the RARA-PML translocation in APL and cells with the RARA SE in AML. The mechanistic studies described here support the therapeutic potential of SY-1425 in myeloid leukemia patients who have a SE associated with RARA. A biomarker directed clinical trial of SY-1425, a potent and selective RARα agonist, in a subset of AML and MDS patients with an altered RARA locus (clinicaltrials.gov, NCT02807558) is supported by these data. Disclosures Fiore: Syros Pharmaceuticals: Employment, Equity Ownership. McKeown:Syros Pharmaceuticals: Employment, Equity Ownership. Lee:Syros Pharmaceuticals: Employment, Equity Ownership. Eaton:Syros Pharmaceuticals: Employment, Equity Ownership. Fritz:Syros Pharmaceuticals: Employment, Equity Ownership.
- Published
- 2016
23. Surface Replacement Hemiarthroplasty for the Treatment of Osteonecrosis of the Femoral Head*
- Author
-
Christopher Fiore, Kenneth A. Krackow, David S. Hungerford, Richard D. Scott, Michael A. Mont, and Marc W. Hungerford
- Subjects
Adult ,Male ,musculoskeletal diseases ,medicine.medical_specialty ,Bone disease ,Arthroplasty, Replacement, Hip ,medicine.medical_treatment ,Prosthesis Design ,Prosthesis ,Arthroplasty ,Femoral head ,Femur Head Necrosis ,medicine ,Humans ,Orthopedics and Sports Medicine ,Femur ,Aged ,business.industry ,Femur Head ,General Medicine ,Perioperative ,Middle Aged ,medicine.disease ,Surgery ,Radiography ,medicine.anatomical_structure ,Harris Hip Score ,Orthopedic surgery ,Female ,business ,Follow-Up Studies - Abstract
We reviewed the results of thirty-three femoral resurfacing procedures in twenty-five patients who had stage-III or early stage-IV osteonecrosis of the femoral head according to the classification system of Ficat and Arlet. There were no perioperative complications. Thirty hip prostheses (91 percent) survived for a minimum of five years. At a mean of 10.5 years (range, four to fourteen years) postoperatively, sixteen (62 percent) of the twenty-six hips with stage-III disease had a good or excellent Harris hip score. Four of the seven hips with stage-IV disease did not have or need a total hip arthroplasty. Overall, twenty hips (61 percent) had a good or excellent result according to the scoring system of Harris, and thirteen (39 percent) had a fair or poor result and subsequently had or needed a total hip arthroplasty. The mean interval between the hemiarthroplasty and the total hip arthroplasty was sixty months (range, thirty-six to 136 months). These thirteen hips all had a successful clinical result (a Harris hip score of at least 80 points) at a mean of thirty months (range, twenty-four to seventy-two months) after the total hip arthroplasty. The results of the present study suggest that resurfacing of the femoral head can be a successful interim procedure for the management of patients who have Ficat and Arlet stage-III or early stage-IV disease with a large lesion that is not amenable to other treatment options except total hip arthroplasty.
- Published
- 1998
24. Utility of multispectral imaging in automated quantitative scoring of immunohistochemistry
- Author
-
Stephen P. Finn, Neil E. Martin, Michelangelo Fiorentino, Swen-Olof Andersson, Dyane Bailey, Ove Andrén, Katja Fall, Niamh Conlon, Massimo Loda, Richard Flavin, Christopher Fiore, Xiaoqiu Wu, Fiore C, Bailey D, Conlon N, Wu X, Martin N, Fiorentino M, Finn S, Fall K, Andersson SO, Andren O, Loda M, and Flavin R
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Cytoplasm ,Multispectral image ,Adenocarcinoma ,Article ,Pathology and Forensic Medicine ,Software ,Urological cancer ,Biomarkers, Tumor ,Image Processing, Computer-Assisted ,Medicine ,Humans ,Image analysis ,Aged ,Aged, 80 and over ,Cell Nucleus ,business.industry ,Cell Membrane ,Prostatic Neoplasms ,Reproducibility of Results ,Pattern recognition ,General Medicine ,Middle Aged ,Immunohistochemistry ,Ki-67 Antigen ,Prostate cancer, digital imaging ,Stathmin ,Artificial intelligence ,business ,alpha Catenin - Abstract
BACKGROUND: Automated scanning devices and image analysis software provide a means to overcome the limitations of manual semiquantitative scoring of immunohistochemistry. Common drawbacks to automated imaging systems include an inability to classify tissue type and an inability to segregate cytoplasmic and nuclear staining. METHODS: Immunohistochemistry for the membranous marker α-catenin, the cytoplasmic marker stathmin and the nuclear marker Ki-67 was performed on tissue microarrays (TMA) of archival formalin-fixed paraffin-embedded tissue comprising 471 (α-catenin and stathmin) and 511 (Ki-67) cases of prostate adenocarcinoma. These TMA were quantitatively analysed using two commercially available automated image analysers, the Ariol SL-50 system and the Nuance system from CRi. Both systems use brightfield microscopy for automated, unbiased and standardised quantification of immunohistochemistry, while the Nuance system has spectral deconvolution capabilities. RESULTS: Overall concordance between scores from both systems was excellent (r=0.90; 0.83-0.95). The software associated with the multispectral imager allowed accurate automated classification of tissue type into epithelial glandular structures and stroma, and a single-step segmentation of staining into cytoplasmic or nuclear compartments allowing independent evaluation of these areas. The Nuance system, however, was not able to distinguish reliably between tumour and non-tumour tissue. In addition, variance in the labour and time required for analysis between the two systems was also noted. CONCLUSION: Despite limitations, this study suggests some beneficial role for the use of a multispectral imaging system in automated analysis of immunohistochemistry.
- Published
- 2012
25. Examiners: Large and Small
- Author
-
Jonathan C. Lipson, Christopher Fiore Marotta, and Elizabeth Berg
- Published
- 2012
26. Immunohistochemical expression of BRCA1 and lethal prostate cancer
- Author
-
Philip W. Kantoff, Neil E. Martin, Meir J. Stampfer, Stephen P. Finn, Jennifer R. Stark, Gregory L Judson, Richard Flavin, Lorelei A. Mucci, Howard D. Sesso, Michelangelo Fiorentino, Edward Giovannucci, Jennifer A. Sinnott, Christopher Fiore, Massimo Loda, Kathryn L. Penney, Katja Fall, Jing Ma, Fiorentino M, Judson G, Penney K, Flavin R, Stark J, Fiore C, Fall K, Martin N, Ma J, Sinnott J, Giovannucci E, Stampfer M, Sesso HD, Kantoff PW, Finn S, Loda M, and Mucci L
- Subjects
PCA3 ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Tumor suppressor gene ,Proliferation index ,endocrine system diseases ,Genes, BRCA1 ,Biology ,Article ,Prostate cancer ,Prostate ,Cancer stem cell ,medicine ,Biomarkers, Tumor ,Humans ,Prospective Studies ,Neoplasm Metastasis ,skin and connective tissue diseases ,Aged ,Oligonucleotide Array Sequence Analysis ,BRCA1 Protein ,Cancer ,Prostatic Neoplasms ,Cell cycle ,medicine.disease ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Ki-67 Antigen ,Oncology ,Mutation ,Cancer research ,Disease Progression ,BRCA1 prostate cancer prognosis - Abstract
BRCA1 functions as a tumor suppressor; recent work suggests that BRCA1 may also induce cell cycle arrest to allow for DNA repair. We hypothesized that BRCA1 expression in prostate tumor tissue may be associated with prostate cancer progression through regulation of the cell cycle. We used immunohistochemistry to evaluate BRCA1 protein expression in archival tumor samples from 393 prostate cancer cases in the Physicians' Health Study. The men were followed prospectively from diagnosis to development of metastases and mortality. Fifteen percent of tumors stained positive for BRCA1. BRCA1-positive tumors had substantially increased tumor proliferation index compared with negative tumors (47.0 Ki67-positive nuclei versus 10.3, P = 0.0016) and were more likely to develop lethal cancer compared with BRCA1-negative tumors (hazard ratio, 4.6; 95% confidence interval, 2.4–8.7). These findings strengthen the hypothesis that BRCA1 plays a role in cell cycle control and show that BRCA1 is a marker of clinical prostate cancer prognosis. Cancer Res; 70(8); 3136–9. ©2010 AACR.
- Published
- 2010
27. Examining Success
- Author
-
Lipson, Jonathan C., primary and Marotta, Christopher Fiore, additional
- Published
- 2015
- Full Text
- View/download PDF
28. Fatty acid synthase: a metabolic enzyme and candidate oncogene in prostate cancer
- Author
-
Jing Ma, Lorelei A. Mucci, Stacey Ruiz, Chiara Grisanzio, Giorgia Zadra, Toshiro Migita, Stephen P. Finn, Eyoung Shin, Carmen Priolo, Michelangelo Fiorentino, Fumika Inazuka, Meir J. Stampfer, Andrew L. Kung, Sabina Signoretti, Phillip G. Febbo, Emanuele Palescandolo, Wanling Xie, Alessandro Fornari, William C. Hahn, Christopher Fiore, Massimo Loda, Aravind Subramanian, Migita T, Ruiz S, Fornari A, Fiorentino M, Priolo C, Zadra G, Inazuka F, Grisanzio C, Palescandolo E, Shin E, Fiore C, Xie W, Kung AL, Febbo PG, Subramanian A, Mucci L, Ma J, Signoretti S, Stampfer M, Hahn WC, Finn S, and Loda M
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Immunoblotting ,Transplantation, Heterologous ,Apoptosis ,Mice, Transgenic ,Biology ,Adenocarcinoma ,medicine.disease_cause ,Gene Expression Regulation, Enzymologic ,Prostate cancer ,Mice ,Prostate ,Internal medicine ,Cell Line, Tumor ,LNCaP ,medicine ,Animals ,Humans ,Gene Silencing ,RNA, Small Interfering ,Oncogene ,Cancer ,Prostatic Neoplasms ,Oncogenes ,Articles ,medicine.disease ,Flow Cytometry ,Immunohistochemistry ,Up-Regulation ,Fatty Acid Synthase, Type I ,Gene Expression Regulation, Neoplastic ,Fatty acid synthase ,Disease Models, Animal ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Bromodeoxyuridine ,FASN, prostate cancer ,Cancer research ,biology.protein ,Carcinogenesis ,Orchiectomy - Abstract
BACKGROUND: Overexpression of the fatty acid synthase (FASN) gene has been implicated in prostate carcinogenesis. We sought to directly assess the oncogenic potential of FASN. METHODS: We used immortalized human prostate epithelial cells (iPrECs), androgen receptor-overexpressing iPrECs (AR-iPrEC), and human prostate adenocarcinoma LNCaP cells that stably overexpressed FASN for cell proliferation assays, soft agar assays, and tests of tumor formation in immunodeficient mice. Transgenic mice expressing FASN in the prostate were generated to assess the effects of FASN on prostate histology. Apoptosis was evaluated by Hoechst 33342 staining and by fluorescence-activated cell sorting in iPrEC-FASN cells treated with stimulators of the intrinsic and extrinsic pathways of apoptosis (ie, camptothecin and anti-Fas antibody, respectively) or with a small interfering RNA (siRNA) targeting FASN. FASN expression was compared with the apoptotic index assessed by the terminal deoxynucleotidyltransferase-mediated UTP end-labeling method in 745 human prostate cancer samples by using the least squares means procedure. All statistical tests were two-sided. RESULTS: Forced expression of FASN in iPrECs, AR-iPrECs, and LNCaP cells increased cell proliferation and soft agar growth. iPrECs that expressed both FASN and androgen receptor (AR) formed invasive adenocarcinomas in immunodeficient mice (12 of 14 mice injected formed tumors vs 0 of 14 mice injected with AR-iPrEC expressing empty vector (P < .001, Fisher exact test); however, iPrECs that expressed only FASN did not. Transgenic expression of FASN in mice resulted in prostate intraepithelial neoplasia, the incidence of which increased from 10% in 8- to 16-week-old mice to 44% in mice aged 7 months or more (P = .0028, Fisher exact test), but not in invasive tumors. In LNCaP cells, siRNA-mediated silencing of FASN resulted in apoptosis. FASN overexpression protected iPrECs from apoptosis induced by camptothecin but did not protect iPrECs from Fas receptor-induced apoptosis. In human prostate cancer specimens, FASN expression was inversely associated with the apoptotic rate (mean percentage of apoptotic cells, lowest vs highest quartile of FASN expression: 2.76 vs 1.34, difference = 1.41, 95% confidence interval = 0.45 to 2.39, Ptrend = .0046). CONCLUSIONS: These observations suggest that FASN can act as a prostate cancer oncogene in the presence of AR and that FASN exerts its oncogenic effect by inhibiting the intrinsic pathway of apoptosis.
- Published
- 2009
29. Overexpression of fatty acid synthase is associated with palmitoylation of Wnt1 and cytoplasmic stabilization of beta-catenin in prostate cancer
- Author
-
Chandan Sharma, Martin E. Hemler, Lorelei A. Mucci, Raffaella Zamponi, Emanuele Palescandolo, Edward Giovannucci, Carmen Priolo, Massimo Loda, Michelangelo Fiorentino, Christopher Fiore, Stephen P. Finn, Giorgia Zadra, Dolores Di Vizio, Paul L. Nguyen, Dyane Bailey, Wanling Xie, Toshiro Migita, Giuseppe Fedele, Fiorentino M, Zadra G, Palescandolo E, Fedele G, Bailey D, Fiore C, Nguyen PL, Migita T, Zamponi R, Di Vizio D, Priolo C, Sharma C, Xie W, Hemler ME, Mucci L, Giovannucci E, Finn S, and Loda M
- Subjects
Male ,medicine.medical_specialty ,Cytoplasm ,Beta-catenin ,Palmitic Acid ,Mice, Nude ,Wnt1 Protein ,Article ,Pathology and Forensic Medicine ,Prostate cancer ,Mice ,Fatty acid synthase prostate cancer ,Palmitoylation ,Internal medicine ,medicine ,Animals ,Humans ,Molecular Biology ,beta Catenin ,Cell Line, Transformed ,DNA Primers ,Gene knockdown ,biology ,Base Sequence ,Cancer ,Prostatic Neoplasms ,Cell Biology ,medicine.disease ,Immunohistochemistry ,Fatty acid synthase ,Endocrinology ,Apoptosis ,Catenin ,Cancer research ,biology.protein ,RNA Interference ,Fatty Acid Synthases - Abstract
Fatty acid synthase (FASN), a key metabolic enzyme for liponeogenesis highly expressed in several human cancers, displays oncogenic properties such as resistance to apoptosis and induction of proliferation when overexpressed. To date, no mechanism has been identified to explain the oncogenicity of FASN in prostate cancer. We generated immortalized prostate epithelial cells (iPrECs) overexpressing FASN, and found that (14)C-acetate incorporation into palmitate synthesized de novo by FASN was significantly elevated in immunoprecipitated Wnt-1 when compared to isogenic cells not overexpressing FASN. Overexpression of FASN caused membranous and cytoplasmic beta-catenin protein accumulation and activation, whereas FASN knockdown by short-hairpin RNA resulted in a reduction in the extent of beta-catenin activation. Orthotopic transplantation of iPrECs overexpressing FASN in nude mice resulted in invasive tumors that overexpressed beta-catenin. A strong significant association between FASN and cytoplasmic (stabilized) beta-catenin immunostaining was found in 862 cases of human prostate cancer after computerized subtraction of the membranous beta-catenin signal (P
- Published
- 2008
30. Examiners: Large and Small
- Author
-
Lipson, Jonathan C., primary, Marotta, Christopher Fiore, additional, and Berg, Elizabeth, additional
- Published
- 2012
- Full Text
- View/download PDF
31. Abstract PR-01: Vitamin D receptor expression is inversely associated with prostate cancer progression
- Author
-
Massimo Loda, Rosina T. Lis, Jing Ma, Lorelei A. Mucci, Meir J. Stampfer, Julie L. Kasperzyk, Whitney K. Hendrickson, Richard Flavin, Michelangelo Fiorentino, Christopher Fiore, Edward Giovannucci, and Kathryn L. Penney
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Estrogen receptor ,Cancer ,medicine.disease ,Calcitriol receptor ,Androgen receptor ,Prostate-specific antigen ,Prostate cancer ,Endocrinology ,Internal medicine ,medicine ,Vitamin D and neurology ,business ,Estrogen receptor alpha - Abstract
Background: Vitamin D is inversely associated with risk of several malignancies. Circulating vitamin D interacts with the vitamin D receptor (VDR) at the cellular level to inhibit proliferation, increase apoptosis and decrease angiogenesis. Thus, although vitamin D levels appear to be unrelated to total prostate cancer incidence, VDR levels in tumor tissue may influence prostate cancer prognosis. Methods: We examined the immunohistochemical expression of VDR on archival tumor tissue from 841 prostate cancer cases diagnosed between 1982 and 2004 within two ongoing, prospective cohorts: the Physicians' Health Study and Health Professionals Follow-up Study. VDR expression was measured quantitatively using the CRi Vectra™ system. We used Cox proportional hazards regression to estimate hazard ratios (HR) and 95% confidence intervals (CI) for the association of VDR expression with lethal prostate cancer (N=73) through 2008. On a subset of cases, we also examined correlation of tumor VDR expression with circulating 25(OH)D3 and 1alpha,25(OH)2D3 measured at baseline (N=84) in 1982 and two SNPs in VDR: Fok1 and bsm1 (N=140). Results: Men with high tumor VDR expression had significantly lower Gleason score, lower prostate specific antigen (PSA) levels at diagnosis, and were less likely to have advanced tumor stage (p=0.008, p=0.012, p=0.001, respectively). Compared to the lowest quartile, men in the highest quartile of VDR expression were at significantly lower risk of developing lethal prostate cancer (age-adjusted HRs across quartiles Q2=0.95, 95% CI: 0.51–1.79; Q3=0.93, 95% CI: 0.49–1.76; Q4 = 0.23, 95% CI:0.09–0.55). This association was attenuated (HRQ4vsQ1 = 0.42, 95% CI: 0.16–1.09) after further adjustment for pathological tumor stage, Gleason grade, and PSA level at diagnosis. Tumors expressing high levels of VDR had modest downregulation of cell proliferation as measured by Ki67 (r=-0.11). Moreover, expression of estrogen receptor alpha (r=0.40, p Conclusion: In this large prospective study, men with tumors that demonstrated upregulation of VDR had significantly improved clinical features and reduced risk of lethal prostate cancer. In line with experimental studies, the positive correlation between VDR expression, and androgen receptor and estrogen receptor provides evidence that that VDR acts in an androgen- and/or estrogen-dependent manner. These data highlight the potential role of the vitamin D system in preventing prostate cancer progression. Citation Information: Cancer Prev Res 2010;3(1 Suppl):PR-01.
- Published
- 2010
32. Utility of multispectral imaging in automated quantitative scoring of immunohistochemistry.
- Author
-
Christopher Fiore, Dyane Bailey, Niamh Conlon, Xiaoqiu Wu, Neil Martin, Michelangelo Fiorentino, Stephen Finn, Katja Fall, Swen-Olof Andersson, Ove Andren, Massimo Loda, and Richard Flavin
- Abstract
BackgroundAutomated scanning devices and image analysis software provide a means to overcome the limitations of manual semiquantitative scoring of immunohistochemistry. Common drawbacks to automated imaging systems include an inability to classify tissue type and an inability to segregate cytoplasmic and nuclear staining.MethodsImmunohistochemistry for the membranous marker α-catenin, the cytoplasmic marker stathmin and the nuclear marker Ki-67 was performed on tissue microarrays (TMA) of archival formalin-fixed paraffin-embedded tissue comprising 471 (α-catenin and stathmin) and 511 (Ki-67) cases of prostate adenocarcinoma. These TMA were quantitatively analysed using two commercially available automated image analysers, the Ariol SL-50 system and the Nuance system from CRi. Both systems use brightfield microscopy for automated, unbiased and standardised quantification of immunohistochemistry, while the Nuance system has spectral deconvolution capabilities.ResultsOverall concordance between scores from both systems was excellent (r=0.90; 0.83â0.95). The software associated with the multispectral imager allowed accurate automated classification of tissue type into epithelial glandular structures and stroma, and a single-step segmentation of staining into cytoplasmic or nuclear compartments allowing independent evaluation of these areas. The Nuance system, however, was not able to distinguish reliably between tumour and non-tumour tissue. In addition, variance in the labour and time required for analysis between the two systems was also noted.ConclusionDespite limitations, this study suggests some beneficial role for the use of a multispectral imaging system in automated analysis of immunohistochemistry. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
33. Androgen Receptor Regulates a Distinct Transcription Program in Androgen-Independent Prostate Cancer
- Author
-
Jindan Yu, Jason S. Carroll, Meredith M. Regan, Xin Yuan, Philip W. Kantoff, Michelangelo Fiorentino, Clifford A. Meyer, Arul M. Chinnaiyan, Bo Han, Tao Wu, Hongyun Wang, Arjun K. Manrai, Mathieu Lupien, Lawrence D. True, Myles Brown, X. Shirley Liu, Rameen Beroukhim, Qianben Wang, Massimo Loda, Olli A. Jänne, Christopher Fiore, Zhong Chen, Kexin Xu, Yong Zhang, Steven P. Balk, Mark A. Rubin, Wei Li, Rohit Mehra, Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, Chen Z, Beroukhim R, Wang H, Lupien M, Wu T, Regan MM, Meyer CA, Carroll JS, Manrai AK, Jänne OA, Balk SP, Mehra R, Han B, Chinnaiyan AM, Rubin MA, True L, Fiorentino M, Fiore C, Loda M, Kantoff PW, Liu XS, and Brown M
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Male ,Transcriptional Activation ,medicine.medical_specialty ,HUMDISEASE ,Biology ,urologic and male genital diseases ,General Biochemistry, Genetics and Molecular Biology ,Article ,Histones ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Epigenetics ,Enhancer ,Transcription factor ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Biochemistry, Genetics and Molecular Biology(all) ,Prostatic Neoplasms ,medicine.disease ,androgen receptor prostate cancer ,Androgen receptor ,Gene Expression Regulation, Neoplastic ,Endocrinology ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Cancer cell ,Ubiquitin-Conjugating Enzymes ,Cancer research ,Androgens ,FOXA1 ,Cell Division - Abstract
SummaryThe evolution of prostate cancer from an androgen-dependent state to one that is androgen-independent marks its lethal progression. The androgen receptor (AR) is essential in both, though its function in androgen-independent cancers is poorly understood. We have defined the direct AR-dependent target genes in both androgen-dependent and -independent cancer cells by generating AR-dependent gene expression profiles and AR cistromes. In contrast to what is found in androgen-dependent cells, AR selectively upregulates M-phase cell-cycle genes in androgen-independent cells, including UBE2C, a gene that inactivates the M-phase checkpoint. We find that epigenetic marks at the UBE2C enhancer, notably histone H3K4 methylation and FoxA1 transcription factor binding, are present in androgen-independent cells and direct AR-enhancer binding and UBE2C activation. Thus, the role of AR in androgen-independent cancer cells is not to direct the androgen-dependent gene expression program without androgen, but rather to execute a distinct program resulting in androgen-independent growth.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.