The Multi-Kinase Inhibitor Debio 0617B Reduces Maintenance and Self-Renewal of Primary Human AML CD34 Stem/Progenitor Cells

Total Page:16

File Type:pdf, Size:1020Kb

The Multi-Kinase Inhibitor Debio 0617B Reduces Maintenance and Self-Renewal of Primary Human AML CD34 Stem/Progenitor Cells Published OnlineFirst May 3, 2017; DOI: 10.1158/1535-7163.MCT-16-0889 Small Molecule Therapeutics Molecular Cancer Therapeutics The Multi-kinase Inhibitor Debio 0617B Reduces Maintenance and Self-renewal of Primary Human AML CD34þ Stem/Progenitor Cells Maximilien Murone1, Ramin Radpour2, Antoine Attinger1, Anne Vaslin Chessex1, Anne-Laure Huguenin2, Christian M. Schurch€ 3, Yara Banz3, Saumitra Sengupta4, Michel Aguet5, Stefania Rigotti1, Yogeshwar Bachhav1,Fred eric Massiere 1, Murali Ramachandra4, Andres McAllister1, and Carsten Riether2,6 Abstract Acute myelogenous leukemia (AML) is initiated and main- Furthermore, phospho-STAT5 (pSTAT5) signaling is increased in þ tained by leukemia stem cells (LSC). LSCs are therapy-resistant, primary CD34 AML stem/progenitors. STAT3/STAT5 activation cause relapse, and represent a major obstacle for the cure of AML. depends on tyrosine phosphorylation, mediated by several Resistance to therapy is often mediated by aberrant tyrosine upstream TKs. Inhibition of single upstream TKs did not eliminate kinase (TK) activation. These TKs primarily activate downstream LSCs. In contrast, the multi-TK inhibitor Debio 0617B reduced þ signaling via STAT3/STAT5. In this study, we analyzed the poten- maintenance and self-renewal of primary human AML CD34 tial to therapeutically target aberrant TK signaling and to eliminate stem/progenitor cells in vitro and in xenotransplantation experi- LSCs via the multi-TK inhibitor Debio 0617B. Debio 0617B has a ments resulting in long-term elimination of human LSCs and unique profile targeting key kinases upstream of STAT3/STAT5 leukemia. Therefore, inhibition of multiple TKs upstream signaling such as JAK, SRC, ABL, and class III/V receptor TKs. We of STAT3/5 may result in sustained therapeutic efficacy of demonstrate that expression of phospho-STAT3 (pSTAT3) in AML targeted therapy in AML and prevent relapses. Mol Cancer Ther; blasts is an independent prognostic factor for overall survival. 16(8); 1497–510. Ó2017 AACR. Introduction they are resistant to existing therapeutic interventions resulting in high rates of relapse (6, 7). As a consequence, LSC numbers or LSC Acute myelogenous leukemia (AML) originates from onco- gene signatures in blasts are negative predictors for survival (8, 9). gene-transformed hematopoietic stem/progenitor cells known as AML pathogenesis is based on serial acquisition and accumu- leukemia stem cells (LSC; ref. 1). LSCs self-renew by symmetric lation of various mutations in transcription factors (TF) as well as cell division or divide asymmetrically into a stem cell and into growth factor receptor tyrosine kinases (RTK), such as FMS-related differentiated cells (2). Phenotypically, human LSCs in AML are a tyrosine kinase 3 (FLT3; refs. 10, 11). STAT proteins are TFs very heterogeneous population (3). Therefore, LSCs are primarily involved in cytokine signaling. STATs are activated through tyro- characterized by their function and capacity to induce leukemia in sine phosphorylation, typically through cytokine receptor–asso- xenotransplantation experiments (1, 4, 5). From a clinical per- ciated Janus kinases (JAK), cytoplasmic tyrosine kinases (TKs; SRC spective, LSCs are of fundamental interest in the therapy of AML as and ABL families of kinases), or subsets of RTKs (12). Cytoplasmic SRC, JAK, and ABL TKs act upstream of STAT family of TFs 1Debiopharm International S.A., Lausanne, Switzerland. 2Tumor Immunology, (12, 13). The STAT3/STAT5 pathway is crucial for oncogenesis Department of Clinical Research, University of Bern, Bern, Switzerland. 3Institute in several cancer entities including leukemia (14–17). An activat- of Pathology, University of Bern, Bern, Switzerland. 4Aurigene Discovery Tech- JAK2V617F fi 5 ing mutation in JAK2 ( ) was identi ed in patients with nologies Limited, Bangalore, Karnataka, India. Swiss Federal Institute of Tech- myeloproliferative neoplasms (18). Constitutive phosphoryla- nology (EPFL), Lausanne, Switzerland. 6Department of Medical Oncology, Inselspital, University Hospital and University of Bern, Bern, Switzerland. tion of JAK2, STAT3, and/or STAT5 has been reported in AML blasts and is associated with a shorter time to relapse and poor Note: Supplementary data for this article are available at Molecular Cancer outcome (19–22). Treatment of primary human AML cells with Therapeutics Online (http://mct.aacrjournals.org/). JAK2 and FLT3 inhibitors has resulted in reduced proliferation M. Murone and R. Radpour contributed equally to this article. (23). Recently, it has been demonstrated that JAK/STAT signaling þ Current address for M. Murone: Cellestia Biotech AG, Hochbergerstrasse 60C, is increased in CD34 AML LSCs compared with normal cord Basel 4057, Switzerland. blood or peripheral blood stem cells, suggesting that JAK/STAT Corresponding Author: Carsten Riether, Department of Medical Oncology, signaling supports AML LSC growth and survival (24). Further- University Hospital and University of Bern, Murtenstrasse 35, Bern 3008, more, pSTAT5 signaling is increased and essential for the main- Switzerland. Phone: 413-1632-0956; Fax: 413-1632-3297; E-mail: tenance of leukemic stem/progenitor cells (25). [email protected] Therefore, targeting key TKs upstream of pSTAT3/pSTAT5 may doi: 10.1158/1535-7163.MCT-16-0889 be a promising strategy to treat AML. A phase II study with the Ó2017 American Association for Cancer Research. JAK2 inhibitor ruxolitinib in relapsed/refractory AML patients www.aacrjournals.org 1497 Downloaded from mct.aacrjournals.org on September 25, 2021. © 2017 American Association for Cancer Research. Published OnlineFirst May 3, 2017; DOI: 10.1158/1535-7163.MCT-16-0889 Murone et al. showed partial clinical efficacy with complete remission (CR) in 3 Experiments were performed one to two times. Details on repli- of 38 patients (26). However, in all patients, leukemia relapsed cates are indicated in the figure legends. Power calculation was not after a short time period, indicating that inhibiting a single performed to predetermine sample size. upstream TK may not be sufficient to eliminate LSCs. We therefore Human xenograft experiments were approved by the local analyzed the potential of combined inhibition of several key TKs experimental animal committee of the Canton of Bern and upstream of STAT3/STAT5 to treat AML. The multi-TK inhibitor performed according to Swiss laws for animal protection. (TKI), Debio 0617B, has a unique profile targeting key kinases Experimental studies using the disseminated MOLM-13-Luc upstream of STAT3/STAT5, including JAK, SRC, ABL, and a subset model were approved by the Ethics Committee for Animal Exper- of RTKs, class III (FLT3, c-KIT, CSF1R, PDGFRa, and PDGFRb) and imentation of Baden-Wuerttemberg. The experimental protocol V (VEGFR1-3) RTKs (27), which are also known to play a role in was registered by the Regierungspr€asidium Freiburg (G-11/11). STAT3 activation (15). So far, Debio 0617B has documented Experimental studies using MV-4-11 rat model were approved efficacy in STAT3-driven solid tumors in vitro and in vivo (27). by the Commission Nationale de l'Experimentation Animale de In this study, we demonstrate that pSTAT5 signaling is France. þ increased in primary AML CD34 stem/progenitor cells com- pared with bulk leukemia blasts and that nuclear pSTAT3 is an AML patients and construction of tissue microarray independent negative prognostic factor in AML. The multi-TKI Case selection and construction of the tissue microarray (TMA) Debio 0617B reduced maintenance and self-renewal of primary used in the current study was described previously (29). Briefly, þ in vitro human CD34 AML stem/progenitor cells and in xeno- bone marrow hematoxylin and eosin (H&E) slides were reviewed transplantation experiments resulting in elimination of human to confirm the diagnosis and to identify the most representative LSCs, without affecting nonmalignant hematopoietic stem/pro- and blast-rich areas. Punches (0.6 mm) of these areas were then genitor cells. removed from the formalin-fixed, paraffin-embedded (FFPE) tissue block using a fully automated arraying device (TMA Grand- Materials and Methods master, 3D Histech), and a TMA block containing 222 cases was assembled. Cell lines NB4, HT-93, and MV-4-11 cells were kindly provided by Prof. Jurg€ Schwaller, Department of Biomedicine, University Hospital Reagents for treatment Basel (Basel, Switzerland) and Prof. Mario Tschan, Institute of The compounds Debio 0617B (27), sunitinib, quizartinib Pathology, University of Bern (Bern, Switzerland), who obtained (AC220), dasatinib, saracatinib, and ruxolitinib as well as the the cell lines from ATCC and DSMZ in 2015 and 2016. Conse- vehicles EAD-Lip1 and Kleptose HPB solution were provided by quently, no additional authentication was performed by the Debiopharm. authors. All cell lines were tested mycoplasma-free. Cell lines were grown in FCS-containing medium recommended by ATCC Kinome scan ¼ (https://www.lgcstandards-atcc.org/?geo_country ch) with Glu- KINOMEscan was performed as described in ref. 30. Com- taMAX supplemented 100 U/mL penicillin, and 100 mg/mL of pound activity is described as %Ctrl. %Ctrl was calculated as fi streptomycin in a humidi ed atmosphere of 95% air and 5% CO2 at 37C. Media were routinely changed every 3 days. test compound signal À positive control signal  100 negative control signal À positive control signal Patients and healthy controls Peripheral blood samples and bone marrow aspirates were fl obtained from untreated AML patients at diagnosis
Recommended publications
  • Stanford Chem-H Presentation (PDF)
    KiNativ® In situ kinase profiling Stanford University ChEM-H confidential @KiNativPlatform Principle of the KiNativ platform • ATP (or ADP) acyl phosphate binds to, and covalently modifies Lysine residues in the active site • Thus, ATP acyl phosphate with a desthiobiotin tag can be used capture and quantitate kinases in a complex lysate Acyl phosphate Desthiobiotin tag ATP 2 ATP acyl phosphate probe covalently modifies kinase in the active site Lysine 2 Lysine 1 3 ATP acyl phosphate probe covalently modifies kinase in the active site Lysine 2 Lysine 1 4 Samples trypsinized, probe-labeled peptides captured with streptavidin, and analyzed by targeted LC-MS2 Identification Quantitation Explicit determination of peptide Integration of signal from MS2 sequence and probe modification site fragment ions from MS2 spectrum 5 Comprehensive Coverage of Protein and Lipid Kinases Protein kinases Atypical kinases Green: Kinases detected on KiNativ Red: Kinases not detected on KiNativ ~80% of known protein and atypical kinases identified on the platform http://www.kinativ.com/coverage/protein-lipid.html 6 Profiling compound(s) on the KiNativ platform Control sample – add probe Sample: Lysate derived from any cell line or tissue from ANY species Treated sample – add inhibitor followed by probe Inhibited kinase Green: Kinases Blue: Probe Gray: Non-kinases Red: Inhibitor 7 Profiling compound(s) on the KiNativ platform Control sample – add probe MS signalMS Sample: Lysate derived from any cell line or tissue from ANY species Treated sample – add inhibitor
    [Show full text]
  • Cancer Subtype Identification Using Somatic Mutation Data
    www.nature.com/bjc ARTICLE Genetics and Genomics Cancer subtype identification using somatic mutation data Marieke Lydia Kuijjer 1,2, Joseph Nathaniel Paulson1,2,3, Peter Salzman4, Wei Ding5 and John Quackenbush1,2,6 BACKGROUND: With the onset of next-generation sequencing technologies, we have made great progress in identifying recurrent mutational drivers of cancer. As cancer tissues are now frequently screened for specific sets of mutations, a large amount of samples has become available for analysis. Classification of patients with similar mutation profiles may help identifying subgroups of patients who might benefit from specific types of treatment. However, classification based on somatic mutations is challenging due to the sparseness and heterogeneity of the data. METHODS: Here we describe a new method to de-sparsify somatic mutation data using biological pathways. We applied this method to 23 cancer types from The Cancer Genome Atlas, including samples from 5805 primary tumours. RESULTS: We show that, for most cancer types, de-sparsified mutation data associate with phenotypic data. We identify poor prognostic subtypes in three cancer types, which are associated with mutations in signal transduction pathways for which targeted treatment options are available. We identify subtype–drug associations for 14 additional subtypes. Finally, we perform a pan-cancer subtyping analysis and identify nine pan-cancer subtypes, which associate with mutations in four overarching sets of biological pathways. CONCLUSIONS: This study is an important step toward understanding mutational patterns in cancer. British Journal of Cancer (2018) 118:1492–1501; https://doi.org/10.1038/s41416-018-0109-7 INTRODUCTION However, subtype classification using somatic mutations in Cancer is a heterogeneous disease that can develop in different cancer is challenging, mainly because the data are very sparse: tissues and cell types.
    [Show full text]
  • Suppression of Signal Transducer and Activator of Transcription 3 Activation by Butein Inhibits Growth of Human Hepatocellular Carcinoma in Vivo
    Author Manuscript Published OnlineFirst on December 3, 2010; DOI: 10.1158/1078-0432.CCR-10-1123 AuthorPublished manuscripts OnlineFirst have been on peer December reviewed and 3, accepted 2010 as for 10.1158/1078-0432.CCR-10-1123 publication but have not yet been edited. Suppression of Signal Transducer and Activator of Transcription 3 Activation by Butein Inhibits Growth of Human Hepatocellular Carcinoma in vivo Peramaiyan Rajendran1, Tina H. Ong2, Luxi Chen1,3, Feng Li1, Muthu K Shanmugam1, Shireen Vali4, Taher Abbasi4, Shweta Kapoor4, Ashish Sharma4, Alan Prem Kumar1,3, Kam M. Hui2,5 Gautam Sethi1,5 1Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, 2Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research National Cancer Centre, Singapore 169610, 3Cancer Science Institute of Singapore, National University of Singapore, and 4Cellworks Group Inc., California 95070; 4Cellworks Research India Pvt. Ltd, Bangalore 560066, India. Running title: Butein inhibits STAT3 signaling in vitro and in vivo in HCC. 5To whom correspondence should be addressed: 1. Dr. Gautam Sethi, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Phone: +65-65163267; Fax: +65- 68737690; Email: [email protected] Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Copyright © 2010 American Association for Cancer Research Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2010 American Association for Cancer Research. Author Manuscript Published OnlineFirst on December 3, 2010; DOI: 10.1158/1078-0432.CCR-10-1123 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
    [Show full text]
  • Download Product Insert (PDF)
    PRODUCT INFORMATION Pacritinib Item No. 16709 CAS Registry No.: 937272-79-2 Formal Name: 11-[2-(1-pyrrolidinyl)ethoxy]-14,19-dioxa- 5,7,27-triazatetracyclo[19.3.1.12,6.18,12] O heptacosa-1(25),2,4,6(27),8,10,12(26), 16E,21,23-decaene Synonym: SB1518 N N H MF: C28H32N4O3 FW: 472.6 N Purity: ≥98% O Stability: ≥2 years at -20°C Supplied as: A crystalline solid N O UV/Vis.: λmax: 285 nm Laboratory Procedures For long term storage, we suggest that pacritinib​ be stored as supplied at -20°C. It should be stable for at least two years. Pacritinib is supplied as a crystalline solid. A stock solution may be made by dissolving the pacritinib in the solvent of choice. Pacritinib is soluble in the organic solvent DMSO, which should be purged with an inert gas, at a concentration of approximately 0.5 mg/ml (slightly warmed). Pacritinib is sparingly soluble in aqueous solutions. To enhance aqueous solubility, dilute the organic solvent solution into aqueous buffers or isotonic saline. If performing biological experiments, ensure the residual amount of organic solvent is insignificant, since organic solvents may have physiological effects at low concentrations. We do not recommend storing the aqueous solution for more than one day. Description FMS-like tyrosine kinase 3 (FLT3) and Janus kinase 2 (JAK2) are tyrosine kinases that mediate cytokine signaling and are frequently mutated in cancers, particularly acute myeloid leukemia.1,2 Pacritinib is an 1 inhibitor of both FLT3 and JAK2 (IC50s = 22 and 23 nM, respectively).
    [Show full text]
  • Biological Pathways and in Vivo Antitumor Activity Induced by Atiprimod in Myeloma
    Leukemia (2007) 21, 2519–2526 & 2007 Nature Publishing Group All rights reserved 0887-6924/07 $30.00 www.nature.com/leu ORIGINAL ARTICLE Biological pathways and in vivo antitumor activity induced by Atiprimod in myeloma P Neri1,2,3, P Tassone1,2,3, M Shammas1, H Yasui2, E Schipani4, RB Batchu1, S Blotta1,2,3, R Prabhala1, L Catley2, M Hamasaki2, T Hideshima2, D Chauhan2, GS Jacob5, D Picker5, S Venuta3, KC Anderson2 and NC Munshi1,2 1Jerome Lipper Multiple Myeloma Center, Department of Adult Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; 2Boston VA Healthcare System, Department of Medicine, Harvard Medical School, MA, USA; 3Department of Experimental and Clinical Medicine, University of ‘Magna Græcia’ and Cancer Center, Catanzaro, Italy; 4Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA and 5Callisto Pharmaceuticals Inc., New York, NY, USA Atiprimod (Atip) is a novel oral agent with anti-inflammatory tion.7,8 Atip inhibits the inflammatory response and preserves properties. Although its in vitro activity and effects on signaling bone integrity in murine models of rheumatoid arthritis (RA),9–12 in multiple myeloma (MM) have been previously reported, here targets macrophages, inhibits phospholipase A and C in rat we investigated its molecular and in vivo effects in MM. Gene 13,14 expression analysis of MM cells identified downregulation of alveolar macrophages and exhibits antiproliferative and 15–17 genes involved in adhesion, cell-signaling, cell cycle and bone antiangiogenic activities in human cancer models. Impor- morphogenetic protein (BMP) pathways and upregulation of tantly, we have previously reported that Atip inhibits MM cell genes implicated in apoptosis and bone development, follow- growth, induces caspase-mediated apoptosis, blocks the phos- ing Atip treatment.
    [Show full text]
  • Classification Decisions Taken by the Harmonized System Committee from the 47Th to 60Th Sessions (2011
    CLASSIFICATION DECISIONS TAKEN BY THE HARMONIZED SYSTEM COMMITTEE FROM THE 47TH TO 60TH SESSIONS (2011 - 2018) WORLD CUSTOMS ORGANIZATION Rue du Marché 30 B-1210 Brussels Belgium November 2011 Copyright © 2011 World Customs Organization. All rights reserved. Requests and inquiries concerning translation, reproduction and adaptation rights should be addressed to [email protected]. D/2011/0448/25 The following list contains the classification decisions (other than those subject to a reservation) taken by the Harmonized System Committee ( 47th Session – March 2011) on specific products, together with their related Harmonized System code numbers and, in certain cases, the classification rationale. Advice Parties seeking to import or export merchandise covered by a decision are advised to verify the implementation of the decision by the importing or exporting country, as the case may be. HS codes Classification No Product description Classification considered rationale 1. Preparation, in the form of a powder, consisting of 92 % sugar, 6 % 2106.90 GRIs 1 and 6 black currant powder, anticaking agent, citric acid and black currant flavouring, put up for retail sale in 32-gram sachets, intended to be consumed as a beverage after mixing with hot water. 2. Vanutide cridificar (INN List 100). 3002.20 3. Certain INN products. Chapters 28, 29 (See “INN List 101” at the end of this publication.) and 30 4. Certain INN products. Chapters 13, 29 (See “INN List 102” at the end of this publication.) and 30 5. Certain INN products. Chapters 28, 29, (See “INN List 103” at the end of this publication.) 30, 35 and 39 6. Re-classification of INN products.
    [Show full text]
  • Tanibirumab (CUI C3490677) Add to Cart
    5/17/2018 NCI Metathesaurus Contains Exact Match Begins With Name Code Property Relationship Source ALL Advanced Search NCIm Version: 201706 Version 2.8 (using LexEVS 6.5) Home | NCIt Hierarchy | Sources | Help Suggest changes to this concept Tanibirumab (CUI C3490677) Add to Cart Table of Contents Terms & Properties Synonym Details Relationships By Source Terms & Properties Concept Unique Identifier (CUI): C3490677 NCI Thesaurus Code: C102877 (see NCI Thesaurus info) Semantic Type: Immunologic Factor Semantic Type: Amino Acid, Peptide, or Protein Semantic Type: Pharmacologic Substance NCIt Definition: A fully human monoclonal antibody targeting the vascular endothelial growth factor receptor 2 (VEGFR2), with potential antiangiogenic activity. Upon administration, tanibirumab specifically binds to VEGFR2, thereby preventing the binding of its ligand VEGF. This may result in the inhibition of tumor angiogenesis and a decrease in tumor nutrient supply. VEGFR2 is a pro-angiogenic growth factor receptor tyrosine kinase expressed by endothelial cells, while VEGF is overexpressed in many tumors and is correlated to tumor progression. PDQ Definition: A fully human monoclonal antibody targeting the vascular endothelial growth factor receptor 2 (VEGFR2), with potential antiangiogenic activity. Upon administration, tanibirumab specifically binds to VEGFR2, thereby preventing the binding of its ligand VEGF. This may result in the inhibition of tumor angiogenesis and a decrease in tumor nutrient supply. VEGFR2 is a pro-angiogenic growth factor receptor
    [Show full text]
  • Cytokine Signaling in Tumor Progression
    Immune Netw. 2017 Aug;17(4):214-227 https://doi.org/10.4110/in.2017.17.4.214 pISSN 1598-2629·eISSN 2092-6685 Review Article Cytokine Signaling in Tumor Progression Myungmi Lee, Inmoo Rhee* Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea Received: Apr 13, 2017 ABSTRACT Revised: Jun 22, 2017 Accepted: Jun 25, 2017 Cytokines are molecules that play critical roles in the regulation of a wide range of normal *Correspondence to functions leading to cellular proliferation, differentiation and survival, as well as in Inmoo Rhee specialized cellular functions enabling host resistance to pathogens. Cytokines released Department of Bioscience and Biotechnology, in response to infection, inflammation or immunity can also inhibit cancer development Sejong University, 209 Neungdong-ro, and progression. The predominant intracellular signaling pathway triggered by cytokines Gwangjin-gu, Seoul 05006, Korea. is the JAK-signal transducer and activator of transcription (STAT) pathway. Knockout mice Tel: +82-2-6935-2432 E-mail: [email protected] and clinical human studies have provided evidence that JAK-STAT proteins regulate the immune system, and maintain immune tolerance and tumor surveillance. Moreover, aberrant Copyright © 2017. The Korean Association of activation of the JAK-STAT pathways plays an undeniable pathogenic role in several types Immunologists of human cancers. Thus, in combination, these observations indicate that the JAK-STAT This is an Open Access article distributed under the terms of the Creative Commons proteins are promising targets for cancer therapy in humans. The data supporting this view Attribution Non-Commercial License (https:// are reviewed herein. creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial Keywords: Cytokine; JAK-STAT; Cancer; Kinase inhibitor use, distribution, and reproduction in any medium, provided the original work is properly cited.
    [Show full text]
  • Investigating the Mechanism of Hepatocellular Carcinoma Progression by Constructing Genetic and Epigenetic Networks Using NGS Data Identification and Big Database Mining Method
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 48 Research Paper Investigating the mechanism of hepatocellular carcinoma progression by constructing genetic and epigenetic networks using NGS data identification and big database mining method Cheng-Wei Li1, Ping-Yao Chang1, Bor-Sen Chen1 1Laboratory of Control and Systems Biology, National Tsing Hua University, Hsinchu, Taiwan Correspondence to: Bor-Sen Chen, email: [email protected] Keywords: DNA methylation, multiple potential drugs, hepatocarcinogenesis, miRNAs, principal network projection Received: February 19, 2016 Accepted: October 26, 2016 Published: November 04, 2016 ABSTRACT The mechanisms leading to the development and progression of hepatocellular carcinoma (HCC) are complicated and regulated genetically and epigenetically. The recent advancement in high-throughput sequencing has facilitated investigations into the role of genetic and epigenetic regulations in hepatocarcinogenesis. Therefore, we used systems biology and big database mining to construct genetic and epigenetic networks (GENs) using the information about mRNA, miRNA, and methylation profiles of HCC patients. Our approach involves analyzing gene regulatory networks (GRNs), protein-protein networks (PPINs), and epigenetic networks at different stages of hepatocarcinogenesis. The core GENs, influencing each stage of HCC, were extracted via principal network projection (PNP). The pathways during different stages of HCC were compared. We observed that extracellular signals were further transduced to
    [Show full text]
  • Spotlight Review
    Leukemia (2009) 23, 10–24 & 2009 Macmillan Publishers Limited All rights reserved 0887-6924/09 $32.00 www.nature.com/leu SPOTLIGHT REVIEW Bone marrow microenvironment and the identification of new targets for myeloma therapy K Podar, D Chauhan and KC Anderson Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics, Dana Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Harvard Medical School, Boston, MA, USA The development of multiple myeloma (MM) is a complex multi- Signaling cascades activated by cytokines, growth factors and/ step process involving both early and late genetic changes in or adhesion in MM cells include the Ras/Raf/MEK/MAPK- the tumor cell as well as selective supportive conditions by the k bone marrow (BM) microenvironment. Indeed, it is now well pathway, PI3K/Akt-pathway, the JAK/Stat3-pathway, the NF B- established that MM cell-induced disruption of the BM homeo- pathway and the Wnt-pathway. Promising intracellular targets stasis between the highly organized cellular and extracellular for novel therapies also include protein kinase C (PKC) and SPOTLIGHT compartments supports MM cell proliferation, survival, migra- heat-shock proteins (HSPs). Moreover, genomic profiling has tion and drug resistance through activation of various signaling now identified additional stage-specific intracellular targets, (for example, PI3K/Akt, JAK/Stat-, Raf/MEK/MAPK-, NFjB- and which are now under investigation as novel potential therapeutic Wnt-) pathways. Based on our enhanced understanding of the 2,4 functional importance of the MM BM microenvironment and its targets. inter-relation with the MM cell resulting in homing, seeding, Cell surface receptors include integrins, cadherins, selectins, proliferation and survival, new molecular targets have been syndecans, and the immunoglobulin superfamily of cell adhe- identified and derived treatment regimens in MM have already sion molecules including syndecan-1 (CD138), H-CAM (CD44), changed fundamentally during recent years.
    [Show full text]
  • Pacritinib Combined with Sirolimus and Low-Dose Tacrolimus for GVHD
    Author Manuscript Published OnlineFirst on March 22, 2021; DOI: 10.1158/1078-0432.CCR-20-4725 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Pidala J et al GVHD prevention with combined JAK2/mTOR inhibition Pacritinib combined with sirolimus and low-dose tacrolimus for GVHD prevention after allogeneic hematopoietic cell transplantation: Preclinical and Phase I trial results Authors: Joseph Pidala1-3, Kelly Walton4, Hany Elmariah1,3, Jongphil Kim5, Asmita Mishra1,3, Nelli Bejanyan1,3, Taiga Nishihori1,3, Farhad Khimani1,3, Lia Perez1,3, Rawan G. Faramand1,3, Marco L. Davila1-3, Michael L. Nieder1,3, Elizabeth M. Sagatys6, Shernan G. Holtan4, Nicholas J. Lawrence7, Harshani R. Lawrence7, Bruce R. Blazar8, Claudio Anasetti1-3, Said M. Sebti9, Brian C. Betts4* Affiliations: 1Department of Blood and Marrow Transplantation – Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA, 2Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA, 3Department of Oncologic Sciences, University of South Florida, Tampa, FL, USA, 4Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA, 5Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA, 6Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, USA, 7Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL, USA, 8Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA, 9Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia USA. Running Title: GVHD prevention with combined JAK2/mTOR inhibition *Address correspondence to: Brian C. Betts, MD Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Nils Hasselmo Hall, Room 2-108, 312 Church Street SE Minneapolis, MN 55455 [email protected] COI statement: B.C.B.
    [Show full text]
  • Comparison of Janus Kinase Inhibitors to Block the Type I Interferon Pathway in Human
    bioRxiv preprint doi: https://doi.org/10.1101/2021.02.08.430317; this version posted February 8, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Title: Comparison of Janus kinase inhibitors to block the type I interferon pathway in human skeletal muscle cells Authors: Travis B. Kinder1* and James Inglese1,2 1National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA, 2National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA *corresponding author: Travis B. Kinder, PhD Post-Doctoral Research Fellow, Assay Development and Screening Technology, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850 [email protected] Funding: This work was supported by a grant from the Cure Juvenile Myositis Foundation and the intramural program of NCATS, NIH, project 1ZIATR000342 (J.I.). Competing interests: None ORCID iD: Travis Kinder: 0000-0003-4161-8213 and James Inglese: 0000-0002-7332-5717 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.08.430317; this version posted February 8, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Abstract The family of Janus kinases (JAK1, JAK2, JAK3, TYK2) mediate signal transduction from cytokine receptors by phosphorylation and activation of intracellular signaling pathways and transcription factors.
    [Show full text]