Full Text (PDF)

Total Page:16

File Type:pdf, Size:1020Kb

Full Text (PDF) Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863 Small Molecule Therapeutics Molecular Cancer Therapeutics Targeting of Hematologic Malignancies with PTC299, A Novel Potent Inhibitor of Dihydroorotate Dehydrogenase with Favorable Pharmaceutical Properties Liangxian Cao1, Marla Weetall1, Christopher Trotta1, Katherine Cintron1, Jiyuan Ma1, Min Jung Kim1, Bansri Furia1, Charles Romfo1, Jason D. Graci1, Wencheng Li1, Joshua Du1, Josephine Sheedy1, Jean Hedrick1, Nicole Risher1, Shirley Yeh1, Hongyan Qi1, Tamil Arasu1, Seongwoo Hwang1,William Lennox1, Ronald Kong1, Janet Petruska1,Young-Choon Moon1, John Babiak1, Thomas W. Davis1, Allan Jacobson2, Neil G. Almstead1, Art Branstrom1, Joseph M. Colacino1, and Stuart W. Peltz1 Abstract PTC299 was identified as an inhibitor of VEGFA mRNA engagement in the DHODH conformation in situ is translation in a phenotypic screen and evaluated in the required for its optimal activity. PTC299 has broad and clinic for treatment of solid tumors. To guide precision potent activity against hematologic cancer cells in preclin- cancer treatment, we performed extensive biological char- ical models, reflecting a reduced pyrimidine nucleotide acterization of the activity of PTC299 and demonstrated salvage pathway in leukemia cells. Archived serum samples that inhibition of VEGF production and cell proliferation from patients treated with PTC299 demonstrated increased by PTC299 is linked to a decrease in uridine nucleotides levels of dihydroorotate, the substrate of DHODH, indi- by targeting dihydroorotate dehydrogenase (DHODH), a cating target engagement in patients. PTC299 has advan- rate-limiting enzyme for de novo pyrimidine nucleotide tages over previously reported DHODH inhibitors, includ- synthesis. Unlike previously reported DHODH inhibitors ing greater potency, good oral bioavailability, and lack of that were identified using in vitro enzyme assays, PTC299 off-target kinase inhibition and myelosuppression, and is a more potent inhibitor of DHODH in isolated mito- thus may be useful for the targeted treatment of hemato- chondria suggesting that mitochondrial membrane lipid logic malignancies. Introduction fourth enzymatic step, the ubiquinone-mediated oxidation of dihydroorotate to orotate in the de novo pyrimidine synthesis De novo pyrimidine nucleotide biosynthesis is activated in pathway (6). DHODH is a validated therapeutic target for the proliferating cancer cells in response to an increased demand for treatment of autoimmune diseases such as rheumatoid arthritis synthesis of DNA, RNA, and phospholipids by activation of (7, 8). Inhibition of DHODH was shown in preclinical studies to oncogenic pathways such as Myc, PI3K, PTEN, and mTOR (1– prevent growth of many types of cancers (9–13). Moreover, it was 5). Dihydroorotate dehydrogenase (DHODH), located on the demonstrated that inhibition of DHODH induces differentiation surface of the inner mitochondrial membrane, catalyzes the of acute myeloid leukemia (AML) cells (14–16). Pharmacologic inhibition of de novo pyrimidine synthesis sensitizes triple-nega- tive breast cancer cells to genotoxic chemotherapy agents and 1PTC Therapeutics, Inc., South Plainfield, New Jersey. 2Department of Microbi- reduces chemotherapy resistance (2). DHODH inhibitors such as ology and Physiological Systems, University of Massachusetts Medical School, teriflunomide, brequinar, and vidofludimus have been consid- Worcester, Massachusetts. ered for use in oncology (17–20), but their use may be limited due Note: Supplementary data for this article are available at Molecular Cancer to a narrow therapeutic window as a result of lesser potency and/ Therapeutics Online (http://mct.aacrjournals.org/). or off-target activities such as their inhibition of kinases (21, 22). Current address for T. Arasu: US Food and Drug Administration, 629 Cranbury There is great interest in the discovery and development of new Road, 2nd Floor, East Brunswick, NJ 08816; and current address for T.W. Davis: DHODH inhibitors for therapeutic uses (23, 24). These efforts PMV Pharmaceuticals Inc., Cedar Brook Corporate Center, 8 Clarke Drive, have mainly relied on in vitro enzyme assays to define and Cranbury Township, NJ 08512. optimize activity, in which the purified enzyme conformation Corresponding Author: Liangxian Cao, PTC Therapeutics, Inc., 100 Corporate may differ from that found in the cells due to lack of membrane fi CT, South Plain eld, NJ 07080. Phone: 908-912-9132; Fax: 908-222-0567; lipids in the assay (25). Recently, it was demonstrated that E-mail: [email protected] DHODH attaches to the inner mitochondrial membrane by doi: 10.1158/1535-7163.MCT-18-0863 binding charged phospholipids which stabilize the flexible sub- Ó2018 American Association for Cancer Research. strate- and drug-binding site, suggesting that the design and www.aacrjournals.org 3 Downloaded from mct.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 23, 2018; DOI: 10.1158/1535-7163.MCT-18-0863 Cao et al. development of novel inhibitors that additionally engage in lipid (Crown Bioscience); otherwise we did not perform any additional interactions may represent a promising approach to targeting cell line authentication in house. The information on the source of DHODH in vivo (26). cells and when they were obtained can be found in Supplemen- PTC299 was identified as an inhibitor of the translation of tary Table S1. VEGFA mRNA using PTC Therapeutics' proprietary GEMS tech- Stable cell lines were generated as reported previously (27). nology phenotypic screening platform (27). PTC299 was in Briefly, plasmid DNA was transfected into HT1080 cells using clinical trials for the treatment of solid tumors (28, 29) and is Fugene-6 transfection reagent (Roche Diagnostics GmbH), fol- currently being reevaluated for use in hematologic cancers. To lowed by selection with 200 mg/mL hygromycin for luciferase- guide safe and effective precision cancer treatment, we performed stable cell lines or 200 mg/mL Zeocin (Invitrogen) for V5 tag stable extensive biochemical characterization of PTC299 and demon- cells in the culture medium. strated that it interacts with and inhibits DHODH, a rate-limiting enzyme in the de novo biosynthesis of pyrimidine nucleotides (5). Western blot analysis Inhibition of VEGFA production by PTC299 is a downstream Western blot was performed as reported previously (27). Anti- effect of inhibiting de novo pyrimidine synthesis as it can be body (C1) specific for VEGFA was purchased from Santa Cruz completely rescued by exogenously added uridine but not by Biotechnologies (1:200 dilution); antibody specific for human other nucleosides. Collectively, PTC299 represents a novel class of b-actin was purchased from Abcam (1:10,000 dilution); antibody potent DHODH inhibitors, and our data demonstrate the poten- specific for V5 tag was purchased from Invitrogen (1:5,000 tial of PTC299 as a treatment option to address unmet needs in dilution). Antibody specific for DHODH was purchased from cancers, such as leukemia and lymphoma, that have lower levels Proteintech (Cat#: 14877-1-AP, 1:500 dilution); and antibody of uridine salvage activity and thus rely on the de novo biosynthesis specific for human prohibitin was purchased from Thermo of pyrimidine nucleotides for survival and rapid proliferation. Scientific (Cat#: PA5-12274 and PA5-14133, 1:1,000 dilution). Immunodetection was done using the corresponding secondary Materials and Methods antibodies conjugated with infrared dyes or horseradish peroxide. The expression levels of proteins were detected with Odyssey General methods (LI-COR) or using enhanced chemiluminescence (Pierce). DNA constructs were generated using standard procedures as reported previously (27). VEGFA 50- and 30-untranslated regions Determination of ELISA EC and cytotoxicity CC values (UTR) were amplified from human genomic DNA (Clontech), 50 50 All ELISAs were performed using commercially available ELISA while the VEGFA165 open reading frame from RNA derived from kits (R&D Systems) according to the manufacturer's instructions. HeLa cells incubated under hypoxia (1% oxygen) was amplified The screen of 240 cell lines was performed at Crown Bioscience. by RT-PCR. DNAs generated with PCR were confirmed by DNA Cells were treated with PTC299 at a series of doses for 72 hours, and sequencing. the inhibition of cell proliferation was determined using a standard PTC299, PTC-371, GSK983, and vidofludimus (4SC-101) were assay, CellTiter-Glo Luminescent Cell Viability Assay (Promega), synthesized at PTC Therapeutics; for details, see the Supplemen- that measures total cellular adenosine triphosphate (ATP) con- tary Materials and Methods in the Supplementary Data File. centrations as an indicator of cell viability. Data generated from Brequinar (SML0113) and teriflunomide (SML0936) were pur- ELISA or cytotoxicity studies were plotted with Prism software. A chased from Sigma, whereas pyrazofurin-MP (P61-B01A) was sigmoidal dose response with a variable slope regression curve was purchased from TriLink Biotechnologies. generated for each compound. Maximal and minimal inhibitions were set at 100% and 0%, respectively, and CC50 values were Cell culture calculated after curve fitting using the Prism software. Tumor cell lines, except for MOLM-13 which were obtained from Dr. Kensuke Kojima in July 2015 (Saga University, Japan) 35S-labeling and immunoprecipitation 0 and Huh-7 which were obtained in February 2001 from the Stuart HT1080 cells harboring
Recommended publications
  • DNA Methylation Analysis Identifies Patterns in Progressive Glioma Grades to Predict Patient Survival
    International Journal of Molecular Sciences Article DNA Methylation Analysis Identifies Patterns in Progressive Glioma Grades to Predict Patient Survival Jingyin Weng and Nicole Salazar * Department of Biology, San Francisco State University, San Francisco, CA 94132, USA; [email protected] * Correspondence: [email protected] Abstract: DNA methylation is an epigenetic change to the genome that impacts gene activities with- out modification to the DNA sequence. Alteration in the methylation pattern is a naturally occurring event throughout the human life cycle which may result in the development of diseases such as cancer. In this study, we analyzed methylation data from The Cancer Genome Atlas, under the Lower-Grade Glioma (LGG) and Glioblastoma Multiforme (GBM) projects, to identify methylation markers that exhibit unique changes in DNA methylation pattern along with tumor grade progression, to predict patient survival. We found ten glioma grade-associated Cytosine-phosphate-Guanine (CpG) sites that targeted four genes (SMOC1, KCNA4, SLC25A21, and UPP1) and the methylation pattern is strongly associated with glioma specific molecular alterations, primarily isocitrate dehydrogenase (IDH) mutation and chromosome 1p/19q codeletion. The ten CpG sites collectively distinguished a cohort of diffuse glioma patients with remarkably poor survival probability. Our study highlights genes (KCNA4 and SLC25A21) that were not previously associated with gliomas to have contributed to the poorer patient outcome. These CpG sites can aid glioma tumor progression monitoring and serve as prognostic markers to identify patients diagnosed with less aggressive and malignant gliomas that exhibit similar survival probability to GBM patients. Keywords: glioma; glioblastoma; DNA methylation; progression; TCGA; WGCNA; prognosis Citation: Weng, J.; Salazar, N.
    [Show full text]
  • CD29 Identifies IFN-Γ–Producing Human CD8+ T Cells With
    + CD29 identifies IFN-γ–producing human CD8 T cells with an increased cytotoxic potential Benoît P. Nicoleta,b, Aurélie Guislaina,b, Floris P. J. van Alphenc, Raquel Gomez-Eerlandd, Ton N. M. Schumacherd, Maartje van den Biggelaarc,e, and Monika C. Wolkersa,b,1 aDepartment of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; bLandsteiner Laboratory, Oncode Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; cDepartment of Research Facilities, Sanquin Research, 1066 CX Amsterdam, The Netherlands; dDivision of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; and eDepartment of Molecular and Cellular Haemostasis, Sanquin Research, 1066 CX Amsterdam, The Netherlands Edited by Anjana Rao, La Jolla Institute for Allergy and Immunology, La Jolla, CA, and approved February 12, 2020 (received for review August 12, 2019) Cytotoxic CD8+ T cells can effectively kill target cells by producing therefore developed a protocol that allowed for efficient iso- cytokines, chemokines, and granzymes. Expression of these effector lation of RNA and protein from fluorescence-activated cell molecules is however highly divergent, and tools that identify and sorting (FACS)-sorted fixed T cells after intracellular cytokine + preselect CD8 T cells with a cytotoxic expression profile are lacking. staining. With this top-down approach, we performed an un- + Human CD8 T cells can be divided into IFN-γ– and IL-2–producing biased RNA-sequencing (RNA-seq) and mass spectrometry cells. Unbiased transcriptomics and proteomics analysis on cytokine- γ– – + + (MS) analyses on IFN- and IL-2 producing primary human producing fixed CD8 T cells revealed that IL-2 cells produce helper + + + CD8 Tcells.
    [Show full text]
  • Protein Interactions in the Cancer Proteome† Cite This: Mol
    Molecular BioSystems View Article Online PAPER View Journal | View Issue Small-molecule binding sites to explore protein– protein interactions in the cancer proteome† Cite this: Mol. BioSyst., 2016, 12,3067 David Xu,ab Shadia I. Jalal,c George W. Sledge Jr.d and Samy O. Meroueh*aef The Cancer Genome Atlas (TCGA) offers an unprecedented opportunity to identify small-molecule binding sites on proteins with overexpressed mRNA levels that correlate with poor survival. Here, we analyze RNA-seq and clinical data for 10 tumor types to identify genes that are both overexpressed and correlate with patient survival. Protein products of these genes were scanned for binding sites that possess shape and physicochemical properties that can accommodate small-molecule probes or therapeutic agents (druggable). These binding sites were classified as enzyme active sites (ENZ), protein–protein interaction sites (PPI), or other sites whose function is unknown (OTH). Interestingly, the overwhelming majority of binding sites were classified as OTH. We find that ENZ, PPI, and OTH binding sites often occurred on the same structure suggesting that many of these OTH cavities can be used for allosteric modulation of Creative Commons Attribution 3.0 Unported Licence. enzyme activity or protein–protein interactions with small molecules. We discovered several ENZ (PYCR1, QPRT,andHSPA6)andPPI(CASC5, ZBTB32,andCSAD) binding sites on proteins that have been seldom explored in cancer. We also found proteins that have been extensively studied in cancer that have not been previously explored with small molecules that harbor ENZ (PKMYT1, STEAP3,andNNMT) and PPI (HNF4A, MEF2B,andCBX2) binding sites. All binding sites were classified by the signaling pathways to Received 29th March 2016, which the protein that harbors them belongs using KEGG.
    [Show full text]
  • A Novel Resveratrol Analog: Its Cell Cycle Inhibitory, Pro-Apoptotic and Anti-Inflammatory Activities on Human Tumor Cells
    A NOVEL RESVERATROL ANALOG : ITS CELL CYCLE INHIBITORY, PRO-APOPTOTIC AND ANTI-INFLAMMATORY ACTIVITIES ON HUMAN TUMOR CELLS A dissertation submitted to Kent State University in partial fulfillment of the requirements for the degree of Doctor of Philosophy by Boren Lin May 2006 Dissertation written by Boren Lin B.S., Tunghai University, 1996 M.S., Kent State University, 2003 Ph. D., Kent State University, 2006 Approved by Dr. Chun-che Tsai , Chair, Doctoral Dissertation Committee Dr. Bryan R. G. Williams , Co-chair, Doctoral Dissertation Committee Dr. Johnnie W. Baker , Members, Doctoral Dissertation Committee Dr. James L. Blank , Dr. Bansidhar Datta , Dr. Gail C. Fraizer , Accepted by Dr. Robert V. Dorman , Director, School of Biomedical Sciences Dr. John R. Stalvey , Dean, College of Arts and Sciences ii TABLE OF CONTENTS LIST OF FIGURES……………………………………………………………….………v LIST OF TABLES……………………………………………………………………….vii ACKNOWLEDGEMENTS….………………………………………………………….viii I INTRODUCTION….………………………………………………….1 Background and Significance……………………………………………………..1 Specific Aims………………………………………………………………………12 II MATERIALS AND METHODS.…………………………………………….16 Cell Culture and Compounds…….……………….…………………………….….16 MTT Cell Viability Assay………………………………………………………….16 Trypan Blue Exclusive Assay……………………………………………………...18 Flow Cytometry for Cell Cycle Analysis……………..……………....……………19 DNA Fragmentation Assay……………………………………………...…………23 Caspase-3 Activity Assay………………………………...……….….…….………24 Annexin V-FITC Staining Assay…………………………………..…...….………28 NF-kappa B p65 Activity Assay……………………………………..………….…29
    [Show full text]
  • A High-Throughput Approach to Uncover Novel Roles of APOBEC2, a Functional Orphan of the AID/APOBEC Family
    Rockefeller University Digital Commons @ RU Student Theses and Dissertations 2018 A High-Throughput Approach to Uncover Novel Roles of APOBEC2, a Functional Orphan of the AID/APOBEC Family Linda Molla Follow this and additional works at: https://digitalcommons.rockefeller.edu/ student_theses_and_dissertations Part of the Life Sciences Commons A HIGH-THROUGHPUT APPROACH TO UNCOVER NOVEL ROLES OF APOBEC2, A FUNCTIONAL ORPHAN OF THE AID/APOBEC FAMILY A Thesis Presented to the Faculty of The Rockefeller University in Partial Fulfillment of the Requirements for the degree of Doctor of Philosophy by Linda Molla June 2018 © Copyright by Linda Molla 2018 A HIGH-THROUGHPUT APPROACH TO UNCOVER NOVEL ROLES OF APOBEC2, A FUNCTIONAL ORPHAN OF THE AID/APOBEC FAMILY Linda Molla, Ph.D. The Rockefeller University 2018 APOBEC2 is a member of the AID/APOBEC cytidine deaminase family of proteins. Unlike most of AID/APOBEC, however, APOBEC2’s function remains elusive. Previous research has implicated APOBEC2 in diverse organisms and cellular processes such as muscle biology (in Mus musculus), regeneration (in Danio rerio), and development (in Xenopus laevis). APOBEC2 has also been implicated in cancer. However the enzymatic activity, substrate or physiological target(s) of APOBEC2 are unknown. For this thesis, I have combined Next Generation Sequencing (NGS) techniques with state-of-the-art molecular biology to determine the physiological targets of APOBEC2. Using a cell culture muscle differentiation system, and RNA sequencing (RNA-Seq) by polyA capture, I demonstrated that unlike the AID/APOBEC family member APOBEC1, APOBEC2 is not an RNA editor. Using the same system combined with enhanced Reduced Representation Bisulfite Sequencing (eRRBS) analyses I showed that, unlike the AID/APOBEC family member AID, APOBEC2 does not act as a 5-methyl-C deaminase.
    [Show full text]
  • Supplementary Table 1: Gene List of 44 Upregulated Enzymes in Transformed Mesenchymal Stem Cell Cancer Model
    Supplementary Table 1: Gene list of 44 upregulated enzymes in transformed mesenchymal stem cell cancer model. Gene expression values for parental MSC (MSC 0) and transformed MSC (MSC5) are an average of three replicate log-2 transformed expression values from affymetrix U133 plus 2 genechip experiments with the log-fold change (LFC) indicating the difference (MSC5-MSC0). Supplementary Table 1 HGNC Symbol Alias Enzyme ID U133 plus2 probe set MSC0 MSC5 LFC Ttest pval # Gene rifs # Pubmed cites from Genecards (Sep 2007) Pathway / Function PharmGKB Drugs? Drug pathways? Therapeutic Target Database Thomson Pharma RNASEH2A AGS4; JUNB; RNHL; RNHIA; RNASEHI 3.1.26.- 203022_at 8.56 9.87 1.31 1.56E-05 0 11 RNA degradation none none none PPAP2C LPP2; PAP-2c; PAP2-g 3.1.3.4 209529_at 6.48 8.63 2.16 5.74E-03 2 13 Glycerolipid synthesis none none none ADARB1 ADAR2, ADAR2a, ADAR2a-L1, ADAR2a-L2, ADAR2a-L3, ADAR2b, ADAR2c 3.5.-.- 234799_at 6.36 8.15 1.79 2.03E-04 10 58 RNA pre-mRNA editing none none none ADARB1 3.5.-.- 203865_s_at 6.99 8.42 1.43 6.94E-03 10 58 RNA pre-mRNA editing none none none UAP1 AgX; AGX1; SPAG2 2.7.7.23 209340_at 11.17 12.45 1.28 2.46E-07 0 37 polysaccharide synthesis none none RNMT MET; RG7MT1; hCMT1c; KIAA0398; DKFZp686H1252 2.1.1.56 202684_s_at 5.70 6.78 1.08 8.15E-03 1 24 RNA (mRNA) capping none none GPD2 GDH2, mGPDH 1.1.1.8 211613_s_at 5.71 6.73 1.02 7.02E-03 2 37 glycolysis none none GCDH ACAD5, GCD 1.3.99.7 237304_at 5.38 6.39 1.01 2.44E-02 4 63 lys, hydroxy-lys, and trp metabolism none none ESPL1 3.4.22.49 38158_at 8.03
    [Show full text]
  • (UPP1) (NM 181597) Human Recombinant Protein Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for TP319125 Uridine Phosphorylase 1 (UPP1) (NM_181597) Human Recombinant Protein Product data: Product Type: Recombinant Proteins Description: Recombinant protein of human uridine phosphorylase 1 (UPP1), transcript variant 2 Species: Human Expression Host: HEK293T Tag: C-Myc/DDK Predicted MW: 33.8 kDa Concentration: >50 ug/mL as determined by microplate BCA method Purity: > 80% as determined by SDS-PAGE and Coomassie blue staining Buffer: 25 mM Tris.HCl, pH 7.3, 100 mM glycine, 10% glycerol Preparation: Recombinant protein was captured through anti-DDK affinity column followed by conventional chromatography steps. Storage: Store at -80°C. Stability: Stable for 12 months from the date of receipt of the product under proper storage and handling conditions. Avoid repeated freeze-thaw cycles. RefSeq: NP_853628 Locus ID: 7378 UniProt ID: Q16831 RefSeq Size: 1857 Cytogenetics: 7p12.3 RefSeq ORF: 930 Synonyms: UDRPASE; UP; UPASE; UPP Summary: This gene encodes a uridine phosphorylase, an enzyme that catalyzes the reversible phosphorylation of uridine (or 2'- deoxyuridine) to uracil and ribose-1-phosphate (or deoxyribose-1-phosphate). The encoded enzyme functions in the degradation and salvage of pyrimidine ribonucleosides. [provided by RefSeq, Oct 2016] Protein Pathways: Drug metabolism - other enzymes, Metabolic pathways, Pyrimidine metabolism This product is to be used for laboratory only. Not for diagnostic or therapeutic use. View online » ©2021 OriGene Technologies, Inc., 9620 Medical Center Drive, Ste 200, Rockville, MD 20850, US 1 / 2 Uridine Phosphorylase 1 (UPP1) (NM_181597) Human Recombinant Protein – TP319125 Product images: Coomassie blue staining of purified UPP1 protein (Cat# TP319125).
    [Show full text]
  • Receptor Signaling Through Osteoclast-Associated Monocyte
    Downloaded from http://www.jimmunol.org/ by guest on September 29, 2021 is online at: average * The Journal of Immunology The Journal of Immunology , 20 of which you can access for free at: 2015; 194:3169-3179; Prepublished online 27 from submission to initial decision 4 weeks from acceptance to publication February 2015; doi: 10.4049/jimmunol.1402800 http://www.jimmunol.org/content/194/7/3169 Collagen Induces Maturation of Human Monocyte-Derived Dendritic Cells by Signaling through Osteoclast-Associated Receptor Heidi S. Schultz, Louise M. Nitze, Louise H. Zeuthen, Pernille Keller, Albrecht Gruhler, Jesper Pass, Jianhe Chen, Li Guo, Andrew J. Fleetwood, John A. Hamilton, Martin W. Berchtold and Svetlana Panina J Immunol cites 43 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Author Choice option Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Freely available online through http://www.jimmunol.org/content/suppl/2015/02/27/jimmunol.140280 0.DCSupplemental This article http://www.jimmunol.org/content/194/7/3169.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Author Choice Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2015 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606.
    [Show full text]
  • Human Breast Cancer Associated Fibroblasts Exhibit Subtype Specific
    Tchou et al. BMC Medical Genomics 2012, 5:39 http://www.biomedcentral.com/1755-8794/5/39 RESEARCH ARTICLE Open Access Human breast cancer associated fibroblasts exhibit subtype specific gene expression profiles Julia Tchou1*, Andrew V Kossenkov2, Lisa Chang2, Celine Satija1, Meenhard Herlyn2, Louise C Showe2† and Ellen Puré2† Abstract Background: Breast cancer is a heterogeneous disease for which prognosis and treatment strategies are largely governed by the receptor status (estrogen, progesterone and Her2) of the tumor cells. Gene expression profiling of whole breast tumors further stratifies breast cancer into several molecular subtypes which also co-segregate with the receptor status of the tumor cells. We postulated that cancer associated fibroblasts (CAFs) within the tumor stroma may exhibit subtype specific gene expression profiles and thus contribute to the biology of the disease in a subtype specific manner. Several studies have reported gene expression profile differences between CAFs and normal breast fibroblasts but in none of these studies were the results stratified based on tumor subtypes. Methods: To address whether gene expression in breast cancer associated fibroblasts varies between breast cancer subtypes, we compared the gene expression profiles of early passage primary CAFs isolated from twenty human breast cancer samples representing three main subtypes; seven ER+, seven triple negative (TNBC) and six Her2+. Results: We observed significant expression differences between CAFs derived from Her2+ breast cancer and CAFs from TNBC and ER + cancers, particularly in pathways associated with cytoskeleton and integrin signaling. In the case of Her2+ breast cancer, the signaling pathways found to be selectively up regulated in CAFs likely contribute to the enhanced migration of breast cancer cells in transwell assays and may contribute to the unfavorable prognosis of Her2+ breast cancer.
    [Show full text]
  • 5-Fluorouracil Enhances the Anti-Tumor Activity of the Glutaminase Inhibitor CB-839 Against
    Author Manuscript Published OnlineFirst on September 9, 2020; DOI: 10.1158/0008-5472.CAN-20-0600 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 5-fluorouracil enhances the anti-tumor activity of the glutaminase inhibitor CB-839 against PIK3CA-mutant colorectal cancers Yiqing Zhao1,2,#, Xiujing Feng1,2,#, Yicheng Chen1,2,#, J. Eva Selfridge1,2,3, Shashank Gorityala4, Zhanwen Du1,2, Janet M. Wang1, Yujun Hao1,2, Gino Cioffi5, Ronald A. Conlon1,2, Jill S. Barnholtz-Sloan2,5, Joel Saltzman3,6, Smitha S. Krishnamurthi3,6, Shaveta Vinayak3,6,7, Martina Veigl2,6, Yan Xu4, David L. Bajor2,3,6, Sanford D. Markowitz2,3,6, Neal J. Meropol2,3,8, Jennifer R. Eads2,3,9*, and Zhenghe Wang1,2,* 1 Department of Genetics and Genome Sciences, 2Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106 3Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 4Department of Chemistry, Cleveland State University, Cleveland, OH 44115 5Department of Population and Quantitative Health Sciences 6Department of Medicine, 7Department of Medicine, University of Washington, Seattle, WA 98195 8Flatiron Health, New York, NY 10013 9Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104 Running title: CB-839 plus 5-FU inhibit PIK3CA-mutant colon cancer growth Key Words: Colorectal cancer; PIK3CA mutation; glutaminase inhibitor; 5-fluorouracil, clinical trial. # These authors contributed equally. *Corresponding authors: Zhenghe Wang, Phone: 216-368-0446, Fax: 216-368-8919, Email: [email protected] (leading contact); Jennifer Eads, Phone: 215-615-1725, Fax: 215-349-8551, Email: [email protected] Conflict of Interest: NJM is currently an employee of Flatiron Health, Inc., an independent subsidiary of the Roche Group.
    [Show full text]
  • Integrated Multi-Omics Analysis of RB-Loss Identifies Widespread
    ARTICLE https://doi.org/10.1038/s42003-021-02495-2 OPEN Integrated multi-omics analysis of RB-loss identifies widespread cellular programming and synthetic weaknesses Swetha Rajasekaran 1,2, Jalal Siddiqui1,2, Jessica Rakijas1,2, Brandon Nicolay3,4,11, Chenyu Lin1,2, Eshan Khan1,2, Rahi Patel1,2, Robert Morris3,4, Emanuel Wyler 5, Myriam Boukhali3,4, Jayashree Balasubramanyam6, R. Ranjith Kumar6, Capucine Van Rechem 7, Christine Vogel8, Sailaja V. Elchuri6, Markus Landthaler 5, ✉ ✉ Benedikt Obermayer5,9,10, Wilhelm Haas3,4, Nicholas Dyson3,4 & Wayne Miles 1,2 Inactivation of RB is one of the hallmarks of cancer, however gaps remain in our under- 1234567890():,; standing of how RB-loss changes human cells. Here we show that pRB-depletion results in cellular reprogramming, we quantitatively measured how RB-depletion altered the tran- scriptional, proteomic and metabolic output of non-tumorigenic RPE1 human cells. These profiles identified widespread changes in metabolic and cell stress response factors pre- viously linked to E2F function. In addition, we find a number of additional pathways that are sensitive to RB-depletion that are not E2F-regulated that may represent compensatory mechanisms to support the growth of RB-depleted cells. To determine whether these molecular changes are also present in RB1−/− tumors, we compared these results to Reti- noblastoma and Small Cell Lung Cancer data, and identified widespread conservation of alterations found in RPE1 cells. To define which of these changes contribute to the growth of cells with de-regulated E2F activity, we assayed how inhibiting or depleting these proteins affected the growth of RB1−/− cells and of Drosophila E2f1-RNAi models in vivo.
    [Show full text]
  • Downloaded from J Immunol Published Online 24 May 2021 Ol.2000624
    NFAT5 Amplifies Antipathogen Responses by Enhancing Chromatin Accessibility, H3K27 Demethylation, and Transcription Factor Recruitment This information is current as of September 28, 2021. Giulia Lunazzi, Maria Buxadé, Marta Riera-Borrull, Laura Higuera, Sarah Bonnin, Hector Huerga Encabo, Silvia Gaggero, Diana Reyes-Garau, Carlos Company, Luca Cozzuto, Julia Ponomarenko, José Aramburu and Cristina López-Rodríguez Downloaded from J Immunol published online 24 May 2021 http://www.jimmunol.org/content/early/2021/05/24/jimmun ol.2000624 http://www.jimmunol.org/ Supplementary http://www.jimmunol.org/content/suppl/2021/05/24/jimmunol.200062 Material 4.DCSupplemental Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 28, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Author Choice Freely available online through The Journal of Immunology Author Choice option Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2021 by The American Association
    [Show full text]