Integrative Epigenomic Analysis Identifies Biomarkers and Therapeutic Targets in Adult B-Acute Lymphoblastic Leukemia

Total Page:16

File Type:pdf, Size:1020Kb

Integrative Epigenomic Analysis Identifies Biomarkers and Therapeutic Targets in Adult B-Acute Lymphoblastic Leukemia Published OnlineFirst October 29, 2012; DOI: 10.1158/2159-8290.CD-12-0208 RESEARCH ARTICLE Integrative Epigenomic Analysis Identifi es Biomarkers and Therapeutic Targets in Adult B-Acute Lymphoblastic Leukemia Huimin Geng 1 , 2 , 8 , Sarah Brennan 1 , Thomas A. Milne 3 , 10 , Wei-Yi Chen 4 , Yushan Li 1 , Christian Hurtz 7 , 8 , 15 , Soo-Mi Kweon 7 , Lynette Zickl 9 , Seyedmehdi Shojaee 7 , 8 , Donna Neuberg 9 , Chuanxin Huang 1 , Debabrata Biswas 4 , Yuan Xin 1 , Janis Racevskis 6 , Rhett P. Ketterling 11 , Selina M. Luger 12 , Hillard Lazarus 14 , Martin S. Tallman 5 , Jacob M. Rowe 13 , Mark R. Litzow 11 , Monica L. Guzman 1 , C. David Allis 3 , Robert G. Roeder 4 , Markus Müschen 7 , 8 , Elisabeth Paietta 6 , Olivier Elemento 2 , and Ari M. Melnick 1 Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst October 29, 2012; DOI: 10.1158/2159-8290.CD-12-0208 ABSTRACT Genetic lesions such as BCR–ABL1 , E2A–PBX1 , and MLL rearrangements (MLLr ) are associated with unfavorable outcomes in adult B-cell precursor acute lymphoblastic leukemia (B-ALL). Leukemia oncoproteins may directly or indirectly disrupt cytosine methylation pat- terning to mediate the malignant phenotype. We postulated that DNA methylation signatures in these aggressive B-ALLs would point toward disease mechanisms and useful biomarkers and therapeutic tar- gets. We therefore conducted DNA methylation and gene expression profi ling on a cohort of 215 adult patients with B-ALL enrolled in a single phase III clinical trial (ECOG E2993) and normal control B cells. In BCR–ABL1 -positive B-ALLs, aberrant cytosine methylation patterning centered around a cytokine network defi ned by hypomethylation and overexpression of IL2RA (CD25). The E2993 trial clinical data showed that CD25 expression was strongly associated with a poor outcome in patients with ALL regard- less of BCR–ABL1 status, suggesting CD25 as a novel prognostic biomarker for risk stratifi cation in B-ALLs. In E2A –PBX1 -positive B-ALLs, aberrant DNA methylation patterning was strongly associated with direct fusion protein binding as shown by the E2A–PBX1 chromatin immunoprecipitation (ChIP) sequencing (ChIP-seq), suggesting that E2A–PBX1 fusion protein directly remodels the epigenome to impose an aggressive B-ALL phenotype. MLL r B-ALL featured prominent cytosine hypomethylation, which was linked with MLL fusion protein binding, H3K79 dimethylation, and transcriptional upregula- tion, affecting a set of known and newly identifi ed MLL fusion direct targets with oncogenic activity such as FLT3 and BCL6 . Notably, BCL6 blockade or loss of function suppressed proliferation and survival of MLL r leukemia cells, suggesting BCL6-targeted therapy as a new therapeutic strategy for MLL r B-ALLs. SIGNIFICANCE: We conducted the fi rst integrative epigenomic study in adult B-ALLs, as a correlative study to the ECOG E2993 phase III clinical trial. This study links for the fi rst time the direct actions of oncogenic fusion proteins with disruption of epigenetic regulation mediated by cytosine methylation. We identify a novel clinically actionable biomarker in B-ALLs: IL2RA (CD25), which is linked with BCR– ABL1 and an infl ammatory signaling network associated with chemotherapy resistance. We show that BCL6 is a novel MLL fusion protein target that is required to maintain the proliferation and survival of primary human adult MLLr cells and provide the basis for a clinical trial with BCL6 inhibitors for patients with MLLr. Cancer Discov; 2(11); 1–20. ©2012 AACR. INTRODUCTION each of these B-ALL subtypes features distinct and per- turbed gene expression profi les as compared with each other Adult B-cell precursor acute lymphoblastic leukemia and to normal pre-B cells ( 3–6 ). A deeper understanding of (B-ALL) is an aggressive disease with a less than 40% long- the mechanisms driving aberrant gene expression as well as term survival rate ( 1 ). This relatively poor outcome compared improved biomarkers and therapeutic targets are needed to with childhood B-ALLs is partly explained by an increased improve risk stratifi cation and therapy. frequency of high-risk molecular lesions such as BCR–ABL1 Transcriptional regulation and hence cellular phenotypes (20%–40% in adults vs. 2%–5% in children), MLL rearrange- are increasingly understood to be programmed by epigenetic ments ( MLLr , 10%–20%), and E2A–PBX1 fusions (5%; refs. 1, modifi cations of chromatin ( 7, 8 ). Epigenetic information is 2 ). The molecular mechanisms underlying poor outcome in encoded, in large part, by patterns of cytosine methylation and these adult B-ALLs are only partially understood. However, histone modifi cations. An increased abundance of cytosine Authors’ Affi liations: 1 Department of Medicine/Hematology-Oncology Pennsylvania; 13 Department of Hematology and BMT, Rambam Medical Division, 2 Institute for Computational Biomedicine, Weill Medical College Center, Haifa, Israel; 14 Department of Medicine, Case Western Reserve of Cornell University; 3 Laboratory of Chromatin Biology and Epigenet- University, Cleveland, Ohio; and 15 Max Planck Institute for Immunobiology, ics, 4 Laboratory of Biochemistry and Molecular Biology, the Rockefeller Freiburg, Germany 5 University; Department of Medicine, Memorial Sloan-Kettering Cancer Note: Supplementary data for this article are available at Cancer Discovery 6 Center, New York; Montefi ore Medical Center, Albert Einstein College of Online (http://cancerdiscovery.aacrjournals.org/). Medicine, Bronx, New York, New York; 7 Children’s Hospital Los Angeles, Corresponding Authors: Ari Melnick, Division of Hematology/Oncology, University of Southern California, Los Angeles; 8 Department of Labo- Department of Medicine, Weill Cornell Medical College, 1300 York Ave, ratory Medicine, University of California San Francisco, San Francisco, New York, NY 10065. Phone: 212-746-7643; Fax: 212-746-8866; E-mail: California; 9 Dana Farber Cancer Institute, Boston, Massachusetts; 10 MRC [email protected] ; Olivier Elemento. E-mail: ole2001@med. Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, cornell.edu; and Elisabeth Paietta. E-mail: [email protected] University of Oxford, Headington, Oxford, United Kingdom; 11 Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minne- doi: 10.1158/2159-8290.CD-12-0208 sota; 12 Abramson Cancer Center, University of Pennsylvania, Philadelphia, © 2012 American Association for Cancer Research. NOVEMBER 2012CANCER DISCOVERY | OF2 Downloaded from cancerdiscovery.aacrjournals.org on September 26, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst October 29, 2012; DOI: 10.1158/2159-8290.CD-12-0208 RESEARCH ARTICLE Geng et al. methylation at gene promoters, especially those contain- RESULTS ing CpG islands, is generally associated with transcriptional Specifi c Promoter DNA Methylation Patterning silencing, whereas decreased cytosine methylation may facili- in Genetically Defi ned B-ALL Subtypes tate transcriptional activation. Importantly, perturbations in cytosine methylation patterning are epigenetically retained We reasoned that cytosine methylation patterning would during cell division, enabling dividing cells to transmit tran- provide biologically and clinically signifi cant information scriptional programming to their progeny ( 7, 8 ). This process about B-ALLs in adult patients. Therefore, we conducted allows cells within tissues including tumors to retain their HELP (HpaII tiny fragment enrichment by ligation-mediated specifi c phenotypes. Along these lines, DNA methylation pat- PCR) DNA methylation assays ( 31 ) on a cohort of 215 newly terning has been shown to shift during normal hematopoiesis diagnosed adult patients with B-ALLs with available diagnos- and is believed to play an essential role in lineage specifi ca- tic specimens enrolled in the ECOG E2993 multicenter phase tion ( 9 ). Accordingly, disruption of the function of DNA III clinical trial (Supplementary Tables S1 and S2 for detailed methyltransferases (DNMT) perturbs normal hematopoiesis patient description). Eighty-three of these patients featured ( 10, 11 ). By the same token, DNA methylation patterning BCR–ABL1 translocations, 7 had E2A–PBX1 fusions, and 28 of terminally differentiated cells must be erased for them to harbored MLL rearrangements. We focused our attention return to a pluripotent and self-renewal state ( 12, 13 ). Similar principally on these B-ALLs because of their defi ned genetic to normal tissues, tumors may be dependent on specifi c DNA background and association with poor clinical outcome. We methylation patterns to acquire their unique phenotypes also profi led normal pre-B cells (CD19+ and VpreB + ) isolated ( 14 ). Characterization of cytosine methylation patterning in from the bone marrows of 12 healthy adults as normal tumors may thus provide important insights into how gene counterpart for our B-ALL tumor samples (Supplementary expression is perturbed in different tumor types. Table S2). HELP was conducted using a customized micro- Identifi cation of epigenetically modifi ed genes may be array design covering more than 50,000 CpGs annotated highly informative, as deregulation of key signaling or tran- to 14,000 gene promoters and our standard quality control scriptional regulatory genes can alter entire downstream and normalization algorithms ( 31–33 ). We conducted tech- pathways and have signifi cant biologic effects. For example, nical validation
Recommended publications
  • REVIEW Signal Transduction, Cell Cycle Regulatory, and Anti
    Leukemia (1999) 13, 1109–1166 1999 Stockton Press All rights reserved 0887-6924/99 $12.00 http://www.stockton-press.co.uk/leu REVIEW Signal transduction, cell cycle regulatory, and anti-apoptotic pathways regulated by IL-3 in hematopoietic cells: possible sites for intervention with anti-neoplastic drugs WL Blalock1, C Weinstein-Oppenheimer1,2, F Chang1, PE Hoyle1, X-Y Wang3, PA Algate4, RA Franklin1,5, SM Oberhaus1,5, LS Steelman1 and JA McCubrey1,5 1Department of Microbiology and Immunology, 5Leo Jenkins Cancer Center, East Carolina University School of Medicine Greenville, NC, USA; 2Escuela de Quı´mica y Farmacia, Facultad de Medicina, Universidad de Valparaiso, Valparaiso, Chile; 3Department of Laboratory Medicine and Pathology, Mayo Clinic and Foundation, Rochester, MN, USA; and 4Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Over the past decade, there has been an exponential increase growth factor), Flt-L (the ligand for the flt2/3 receptor), erythro- in our knowledge of how cytokines regulate signal transduc- poietin (EPO), and others affect the growth and differentiation tion, cell cycle progression, differentiation and apoptosis. Research has focused on different biochemical and genetic of these early hematopoietic precursor cells into cells of the 1–4 aspects of these processes. Initially, cytokines were identified myeloid, lymphoid and erythroid lineages (Table 1). This by clonogenic assays and purified by biochemical techniques. review will concentrate on IL-3 since much of the knowledge This soon led to the molecular cloning of the genes encoding of how cytokines affect cell growth, signal transduction, and the cytokines and their cognate receptors.
    [Show full text]
  • Molecular and Physiological Basis for Hair Loss in Near Naked Hairless and Oak Ridge Rhino-Like Mouse Models: Tracking the Role of the Hairless Gene
    University of Tennessee, Knoxville TRACE: Tennessee Research and Creative Exchange Doctoral Dissertations Graduate School 5-2006 Molecular and Physiological Basis for Hair Loss in Near Naked Hairless and Oak Ridge Rhino-like Mouse Models: Tracking the Role of the Hairless Gene Yutao Liu University of Tennessee - Knoxville Follow this and additional works at: https://trace.tennessee.edu/utk_graddiss Part of the Life Sciences Commons Recommended Citation Liu, Yutao, "Molecular and Physiological Basis for Hair Loss in Near Naked Hairless and Oak Ridge Rhino- like Mouse Models: Tracking the Role of the Hairless Gene. " PhD diss., University of Tennessee, 2006. https://trace.tennessee.edu/utk_graddiss/1824 This Dissertation is brought to you for free and open access by the Graduate School at TRACE: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Doctoral Dissertations by an authorized administrator of TRACE: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. To the Graduate Council: I am submitting herewith a dissertation written by Yutao Liu entitled "Molecular and Physiological Basis for Hair Loss in Near Naked Hairless and Oak Ridge Rhino-like Mouse Models: Tracking the Role of the Hairless Gene." I have examined the final electronic copy of this dissertation for form and content and recommend that it be accepted in partial fulfillment of the requirements for the degree of Doctor of Philosophy, with a major in Life Sciences. Brynn H. Voy, Major Professor We have read this dissertation and recommend its acceptance: Naima Moustaid-Moussa, Yisong Wang, Rogert Hettich Accepted for the Council: Carolyn R.
    [Show full text]
  • AFF3 Upregulation Mediates Tamoxifen Resistance in Breast
    Shi et al. Journal of Experimental & Clinical Cancer Research (2018) 37:254 https://doi.org/10.1186/s13046-018-0928-7 RESEARCH Open Access AFF3 upregulation mediates tamoxifen resistance in breast cancers Yawei Shi1†, Yang Zhao2†, Yunjian Zhang1, NiJiati AiErken3, Nan Shao1, Runyi Ye1, Ying Lin1* and Shenming Wang1* Abstract Background: Although tamoxifen is a highly effective drug for treating estrogen receptor–positive (ER+) breast cancer, nearly all patients with metastasis with initially responsive tumors eventually relapse, and die from acquired drug resistance. Unfortunately, few molecular mediators of tamoxifen resistance have been described. Here, we describe AFF3 (AF4/FMR2 family member 3), which encodes a nuclear protein with transactivation potential that confers tamoxifen resistance and enables estrogen-independent growth. Methods: We investigated AFF3 expression in breast cancer cells and in clinical breast cancer specimens with western blot and Real-time PCR. We also examined the effects of AFF3 knockdown and overexpression on breast cancer cells using luciferase, tetrazolium, colony formation, and anchorage-independent growth assays in vitro and with nude mouse xenografting in vivo. Results: AFF3 was overexpressed in tamoxifen-resistant tumors. AFF3 overexpression in breast cancer cells resulted in tamoxifen resistance, whereas RNA interference–mediated gene knockdown reversed this phenotype. Furthermore, AFF3 upregulation led to estrogen-independent growth in the xenograft assays. Mechanistic investigations revealed that AFF3 overexpression activated the ER signaling pathway and transcriptionally upregulated a subset of ER-regulated genes. Clinical analysis showed that increased AFF3 expression in ER+ breast tumors was associated with worse overall survival. Conclusions: These studies establish AFF3 as a key mediator of estrogen-independent growth and tamoxifen resistance and as a potential novel diagnostic and therapeutic target.
    [Show full text]
  • BCL-2 in the Crosshairs: Tipping the Balance of Life and Death
    Cell Death and Differentiation (2006) 13, 1339–1350 & 2006 Nature Publishing Group All rights reserved 1350-9047/06 $30.00 www.nature.com/cdd Review BCL-2 in the crosshairs: tipping the balance of life and death LD Walensky*,1,2,3 the founding member of a family of proteins that regulate cell death.1–3 Gene rearrangement places BCL-2 under the 1 Departments of Pediatric Oncology and Cancer Biology, Dana-Farber Cancer transcriptional control of the immunoglobulin heavy chain Institute, Harvard Medical School, Boston, MA 02115, USA locus, resulting in high-level BCL-2 expression and pathologic 2 Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Harvard cell survival.4,5 The oncogenic activity of BCL-2 derives from Medical School, Boston, MA 02115, USA 3 Division of Hematology/Oncology, Children’s Hospital Boston, Harvard Medical its ability to block cell death following a wide variety of 6–8 School, Boston, MA 02115, USA stimuli. Transgenic mice bearing a BCL-2-Ig minigene * Corresponding author: LD Walensky, Department of Pediatric Oncology, initially displayed a polyclonal follicular lymphoproliferation Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA. that selectively expanded a small resting IgM/IgD B-cell Tel: þ 617-632-6307; Fax: þ 617-632-6401; population.4,9 These recirculating B cells accumulated be- E-mail: [email protected] cause of an extended survival rather than increased prolifera- Received 21.3.06; revised 04.5.06; accepted 09.5.06; published online 09.6.06 tion. Despite a fourfold increase in resting B cell counts, Edited by C Borner BCL-2-Ig mice were initially quite healthy.
    [Show full text]
  • Cancer Chemotherapy with Peptides and Peptidomimetics Drug and Peptide Based-Vaccines
    International Research Journal of Pharmacy and Medical Sciences ISSN (Online): 2581-3277 Cancer Chemotherapy with Peptides and Peptidomimetics Drug and Peptide Based-Vaccines Jeevan R. Rajguru*1, Sonali A. Nagare2, Ashish A. Gawai3, Amol G. Jadhao4, Mrunal K. Shirsat5 1, 4Master of Pharmacy in Pharmaceutics at Anuradha College of Pharmacy, Chikhli, Dist-Buldana M.S, India 2Department of Pharmaceutical Chemistry Loknete Shri Dadapatil Pharate College of pharmacy, Mandavgan Pharata, Shirur Pune, M.S, India 3Associate Professor, Department of Pharmaceutical Chemistry at Anuradha College of pharmacy, Chikhli, Dist- Buldana M.S, India 5Department of Pharmacognosy and Principal of Loknete Shri Dadapatil Pharate College of pharmacy, Mandavgan Pharata, Shirur Pune, M.S, India Corresponding Author Email ID: jeevanrajguru 97 @ gmail. com Co-Author Email ID: sonalinagare 93 @ gmail. com Abstract— A summary of the current status of the application of peptidomimetics in cancer therapeutics as an alternative to peptide drugs is provided. Only compounds that are used in therapy or at least under clinical trials are discussed, using inhibitors of farnesyltransferase, proteasome and matrix metalloproteinases as examples. The design and synthesis of peptidomimetics are most important because of the dominant position peptide and protein-protein interactions play in molecular recognition and signalling, especially in living systems. The design of peptidomimetics can be viewed from several different perspectives and peptidomimetics can be categorized in a number of different ways. Study of the vast literature would suggest that medicinal and organic chemists, who deal with peptide mimics, utilize these methods in many different ways. Conventional methods used to treat cancer, from non-specific chemotherapy to modern molecularly targeted drugs have generated limited results due to the complexity of the disease as well as lack of molecular classes that can be developed into treatments rapidly, easily and economically.
    [Show full text]
  • Leveraging the Bcl-2 Interactome to Kill Cancer Cells—
    Author Manuscript Published OnlineFirst on April 2, 2015; DOI: 10.1158/1078-0432.CCR-14-0959 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Molecular Pathways: Leveraging the Bcl-2 Interactome to Kill Cancer Cells— Mitochondrial Outer Membrane Permeabilization and Beyond Hetal Brahmbhatt1,2, Sina Oppermann2, Elizabeth J. Osterlund2,3, Brian Leber4, and David W. Andrews1,2,3 1Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada. 2Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada. 3Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada. 4Department of Medicine, McMaster University, Hamilton, Ontario, Canada. Corresponding Author: David W. Andrews, Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada. M4N 3M5. Phone: 416-480-5120; Fax: 416-480-4375; E-mail: [email protected] Running Title: Bcl-2 Proteins as Chemotherapy Targets Disclosure of Potential Conflicts of Interest B. Leber reports receiving speakers bureau honoraria from AMGEN Canada, Bristol-Myers Squibb Canada, Celgene Canada, Novartis Canada, and Pfizer Canada. No potential conflicts of interest were disclosed by the other authors. Downloaded from clincancerres.aacrjournals.org on October 2, 2021. © 2015 American Association for Cancer Research. Author Manuscript Published OnlineFirst on April 2, 2015; DOI: 10.1158/1078-0432.CCR-14-0959 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. ABSTRACT The inhibition of apoptosis enables the survival and proliferation of tumors and contributes to resistance to conventional chemotherapy agents and is therefore a very promising avenue for the development of new agents that will enhance current cancer therapies.
    [Show full text]
  • Peptidomimetic Blockade of MYB in Acute Myeloid Leukemia
    bioRxiv preprint doi: https://doi.org/10.1101/222620; this version posted November 20, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Peptidomimetic blockade of MYB in acute myeloid leukemia 2 3 4 Kavitha Ramaswamy1,2, Lauren Forbes1,7, Gerard Minuesa1, Tatyana Gindin3, Fiona Brown1, 5 Michael Kharas1, Andrei Krivtsov4,6, Scott Armstrong2,4,6, Eric Still1, Elisa de Stanchina5, Birgit 6 Knoechel6, Richard Koche4, Alex Kentsis1,2,7* 7 8 9 1 Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA. 10 2 Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA. 11 3 Department of Pathology and Cell Biology, Columbia University Medical Center and New York 12 Presbyterian Hospital, New York, NY, USA. 13 4 Center for Epigenetics Research, Sloan Kettering Institute, New York, NY, USA. 14 5 Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, 15 USA. 16 6 Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. 17 7 Weill Cornell Medical College, Cornell University, New York, NY, USA. 18 19 * Correspondence should be addressed to A.K. ([email protected]). 1 bioRxiv preprint doi: https://doi.org/10.1101/222620; this version posted November 20, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 20 ABSTRACT 21 22 Aberrant gene expression is a hallmark of acute leukemias. However, therapeutic strategies for 23 its blockade are generally lacking, largely due to the pharmacologic challenges of drugging 24 transcription factors.
    [Show full text]
  • Designing Peptidomimetics
    CORE Metadata, citation and similar papers at core.ac.uk Provided by UPCommons. Portal del coneixement obert de la UPC DESIGNING PEPTIDOMIMETICS Juan J. Perez Dept. of Chemical Engineering ETS d’Enginyeria Industrial Av. Diagonal, 647 08028 Barcelona, Spain 1 Abstract The concept of a peptidomimetic was coined about forty years ago. Since then, an enormous effort and interest has been devoted to mimic the properties of peptides with small molecules or pseudopeptides. The present report aims to review different approaches described in the past to succeed in this goal. Basically, there are two different approaches to design peptidomimetics: a medicinal chemistry approach, where parts of the peptide are successively replaced by non-peptide moieties until getting a non-peptide molecule and a biophysical approach, where a hypothesis of the bioactive form of the peptide is sketched and peptidomimetics are designed based on hanging the appropriate chemical moieties on diverse scaffolds. Although both approaches have been used in the past, the former has been more widely used to design peptidomimetics of secretory peptides, whereas the latter is nowadays getting momentum with the recent interest in designing protein-protein interaction inhibitors. The present report summarizes the relevance of the information gathered from structure-activity studies, together with a short review on the strategies used to design new peptide analogs and surrogates. In a following section there is a short discussion on the characterization of the bioactive conformation of a peptide, to continue describing the process of designing conformationally constrained analogs producing first and second generation peptidomimetics. Finally, there is a section devoted to review the use of organic scaffolds to design peptidomimetics based on the information available on the bioactive conformation of the peptide.
    [Show full text]
  • ASTX660, a Novel Non-Peptidomimetic Antagonist of Ciap1/2 and XIAP, Potently Induces Tnfa-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth George A
    Published OnlineFirst April 25, 2018; DOI: 10.1158/1535-7163.MCT-17-0848 Small Molecule Therapeutics Molecular Cancer Therapeutics ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFa-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth George A. Ward, Edward J. Lewis, Jong Sook Ahn, Christopher N. Johnson, John F. Lyons, Vanessa Martins, Joanne M. Munck, Sharna J. Rich, Tomoko Smyth, Neil T. Thompson, Pamela A. Williams, Nicola E. Wilsher, Nicola G. Wallis, and Gianni Chessari Abstract Because of their roles in the evasion of apoptosis, inhibitor of antagonism of XIAP was demonstrated by measuring its dis- apoptosis proteins (IAP) are considered attractive targets for placement from caspase-9 or SMAC. Compound-induced pro- anticancer therapy. Antagonists of these proteins have the teasomal degradation of cIAP1 and 2, resulting in downstream potential to switch prosurvival signaling pathways in cancer effects of NIK stabilization and activation of noncanonical cells toward cell death. Various SMAC-peptidomimetics with NF-kB signaling, demonstrated cIAP1/2 antagonism. Treatment inherent cIAP selectivity have been tested clinically and dem- withASTX660ledtoTNFa-dependent induction of apoptosis onstrated minimal single-agent efficacy. ASTX660 is a potent, in various cancer cell lines in vitro, whereas dosing in mice non-peptidomimetic antagonist of cIAP1/2 and XIAP, discov- bearing breast and melanoma tumor xenografts inhibited ered using fragment-based drug design. The antagonism of tumor growth. ASTX660 is currently being tested in a phase XIAP and cIAP1 by ASTX660 was demonstrated on purified I–II clinical trial (NCT02503423), and we propose that its proteins, cells, and in vivo in xenograft models.
    [Show full text]
  • Targeted Next-Gen Sequencing for Detecting MLL Gene Fusions in Leukemia
    Author Manuscript Published OnlineFirst on November 13, 2017; DOI: 10.1158/1541-7786.MCR-17-0569 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Targeted Next-Gen Sequencing for Detecting MLL Gene Fusions in Leukemia. Sadia Afrin1, Christine RC Zhang1, Claus Meyer2, Caedyn L. Stinson1, Thy Pham1, Timothy J.C. Bruxner3, Nicola C. Venn4, Toby N. Trahair5, Rosemary Sutton4, Rolf Marschalek2, J. Lynn Fink1* and Andrew S. Moore1,6,7* 1The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia 2Institute of Pharm. Biology/DCAL, Goethe-University, Frankfurt/Main, Germany. 3Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia 4Children's Cancer Institute, University of NSW, Sydney, Australia 5Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia 6Oncology Services Group, Children’s Health Queensland Hospital and Health Service, Brisbane, Australia 7UQ Child Health Research Centre, The University of Queensland, Brisbane, Australia Running title: MLL Fusion Detection by Targeted NGS. *These authors contributed equally to this work. Corresponding author: Andrew S. Moore, The University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent St, Woolloongabba, QLD, Australia 4102; T: +61 (0)7 3443 6954; F: +61 (0)7 3443 6966; [email protected] This work was supported by The Kids’ Cancer Project and the Children’s Hospital Foundation (Queensland). SA and ASM are supported by the Children’s Hospital Foundation (Queensland). Disclosure of Potential Conflicts of Interest: The authors declare no potential conflicts of interest. 1 Downloaded from mcr.aacrjournals.org on October 1, 2021. © 2017 American Association for Cancer Research.
    [Show full text]
  • Stapled Peptides—A Useful Improvement for Peptide-Based Drugs
    molecules Review Stapled Peptides—A Useful Improvement for Peptide-Based Drugs Mattia Moiola, Misal G. Memeo and Paolo Quadrelli * Department of Chemistry, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; [email protected] (M.M.); [email protected] (M.G.M.) * Correspondence: [email protected]; Tel.: +39-0382-987315 Received: 30 July 2019; Accepted: 1 October 2019; Published: 10 October 2019 Abstract: Peptide-based drugs, despite being relegated as niche pharmaceuticals for years, are now capturing more and more attention from the scientific community. The main problem for these kinds of pharmacological compounds was the low degree of cellular uptake, which relegates the application of peptide-drugs to extracellular targets. In recent years, many new techniques have been developed in order to bypass the intrinsic problem of this kind of pharmaceuticals. One of these features is the use of stapled peptides. Stapled peptides consist of peptide chains that bring an external brace that force the peptide structure into an a-helical one. The cross-link is obtained by the linkage of the side chains of opportune-modified amino acids posed at the right distance inside the peptide chain. In this account, we report the main stapling methodologies currently employed or under development and the synthetic pathways involved in the amino acid modifications. Moreover, we report the results of two comparative studies upon different kinds of stapled-peptides, evaluating the properties given from each typology of staple to the target peptide and discussing the best choices for the use of this feature in peptide-drug synthesis. Keywords: stapled peptide; structurally constrained peptide; cellular uptake; helicity; peptide drugs 1.
    [Show full text]
  • Phase I Trial of the Matrix Metalloproteinase Inhibitor Marimastat Combined with Carboplatin and Paclitaxel in Patients with Advanced Non−Small Cell Lung Cancer
    Cancer Therapy: Clinical PhaseITrialoftheMatrixMetalloproteinaseInhibitorMarimastat Combined with Carboplatin and Paclitaxel in Patients with Advanced Non ^ Small Cell Lung Cancer John R. Goffin,1, 2 Ian C. Anderson,1,4 Jeffrey G. Supko,3 Joseph Paul Eder, Jr.,1 Geoffrey I. Shapiro,1 Thomas J. Lynch,3 Margaret Shipp,1 Bruce E. Johnson,1 and Arthur T. Skarin 1 Abstract Purpose: Marimastat is an orally bioavailable inhibitor of matrix metalloproteinases. A phase I study was initiated to determine whether conventional doses of carboplatin and paclitaxel are tolerated when combined with marimastat and to assess the influence of marimastat on paclitaxel pharmacokinetics. Experimental Design: Three dose levels were evaluated. Marimastat (10 or 20 mg oral admin- istration b.i.d.) was administered continuously with paclitaxel (175 or 200 mg/m2 as a 3-hour i.v. infusion) and carboplatin (at a dose providing an area under the free drug plasma concentration- time curve of 7 mg min/mL) administered each 3 weeks. Toxicity and response were evaluated throughout the intended four cycles of combined therapy. The plasma pharmacokinetics of pacli- taxel was determinedin each patient both without concurrent marimastat and after receiving mari- mastat for1week. Results: Twenty-two chemotherapy-naive patients with stage IIIb (27%) or stage IV (73%) non ^ small cell lung cancer were enrolled. Their median age was 56 years (range, 39-73 years), 50% were female, and their performance status (Eastern Cooperative Oncology Group) ranged from0 to 2. Treatmentwas well tolerated, as 18 (82%) of the patients completed all four cycles of chemotherapy without dose-limiting toxicity. Grade 2 musculoskeletal toxicities were reported in 3 of 12 patients receiving marimastat (20 mg b.i.d.).
    [Show full text]