Class Effects of Tyrosine Kinase Inhibitors in the Treatment of Chronic Myeloid Leukemia

Total Page:16

File Type:pdf, Size:1020Kb

Class Effects of Tyrosine Kinase Inhibitors in the Treatment of Chronic Myeloid Leukemia Leukemia (2009) 23, 1698–1707 & 2009 Macmillan Publishers Limited All rights reserved 0887-6924/09 $32.00 www.nature.com/leu SPOTLIGHT REVIEW Class effects of tyrosine kinase inhibitors in the treatment of chronic myeloid leukemia FJ Giles1, M O’Dwyer2 and R Swords1 1Institute for Drug Development, Cancer Therapy and Research Center at the University of Texas Health Science Center at San Antonio, San Antonio, TX, USA and 2Department of Hematology, University College Hospital, Galway, Ireland Tyrosine kinase inhibitors have revolutionized the treatment of during treatment.8–11 Several strategies are available to treat chronic myeloid leukemia (CML), offering patients several imatinib-resistant patients, including imatinib dose escalation, targeted therapeutic options that provide the possibility of sustained remissions and prolonged survival. With the avail- dose adjustment based on pharmacokinetic assessments and the ability of imatinib, nilotinib and dasatinib, physicians must use of novel TKIs (Figure 1, Table 1). weigh the efficacy and safety profile of each agent when Nilotinib and dasatinib are potent inhibitors of BCR-ABL that choosing the best therapeutic option for individual patients. are US Food and Drug Administration approved for the SPOTLIGHT Each agent targets tyrosine kinases within the cell uniquely to treatment of patients resistant to, or intolerant of, prior therapy, cause the desired antiproliferative effect. In addition to inhibit- including imatinib. These second-generation TKIs have shown ing the BCR-ABL kinase, imatinib and nilotinib target the same 12–14 array of other tyrosine kinases, including c-KIT and platelet- efficacy in all imatinib-resistant or -intolerant CML patients. derived growth factor receptor (PDGFR), albeit with differing Thus far, nilotinib and dasatinib appear to be similar with regard 15 potencies. While targeting BCR-ABL with the highest potency to clinical efficacy, although they display differential safety among approved agents in CML, dasatinib also targets a broad profiles.16 Other new kinase inhibitors are in early clinical array of off-target kinases, including SRC family members, trials as alternatives to imatinib therapy, including bosutinib, PDGFR and EPHB4. The differences in kinase inhibition profiles MK-0457 and INNO-406 (Figure 1, Table 1). Larger prospective, among these agents in vitro probably account for the differing clinical safety profiles of these agents. This paper reviews the randomized studies are needed to determine the place of these various kinases inhibited by imatinib, nilotinib and dasatinib, new therapies in the treatment algorithm of CML. However, the and describes the potential impact of kinase inhibition on the potential for increased potency of these drugs could eventually efficacy and safety of each agent. lead to their replacement of imatinib as first-line CML therapy. Leukemia (2009) 23, 1698–1707; doi:10.1038/leu.2009.111; Although these new therapies are already changing the land- published online 28 May 2009 scape of CML therapy, it is important to distinguish the Keywords: CML; imatinib; nilotinib; dasatinib; TKI; BCR-ABL differences between these agents regarding target kinase profile, safety and efficacy. This article will review the similarities and differences in the kinase inhibition profiles of imatinib, nilotinib, dasatinib and newer BCR-ABL kinase inhibitors, with a focus on the safety of each agent. Although no randomized clinical Introduction trials have been conducted comparing nilotinib and dasatinib treatments, one must use the data from the available studies to The introduction of the small tyrosine kinase inhibitor (TKI), make an informed choice between the two agents for patients imatinib, has revolutionized the treatment of chronic myeloid who have become resistant or intolerant to imatinib therapy. leukemia (CML).1 Imatinib is a relatively specific inhibitor The following sections will highlight pre-clinical and clinical of BCR-ABL, the gene product of the t(9;22) chromosomal data to assist in choosing between the currently available translocation event that forms the Philadelphia chromo- second-generation agents. some characteristic of CML and some acute lymphocytic leukemias.2,3 The fusion of BCR-ABL results in the constitutive activity of the ABL tyrosine kinase, which ultimately drives Differential kinase inhibition profiles of TKIs in CML transformation of hematopoietic progenitor cells. treatment Imatinib was approved by the US Food and Drug Adminis- tration in 2002 for the first-line treatment of CML based on Inhibition of the BCR-ABL tyrosine kinase is considered to be the its high cytogenetic and molecular response rates, long-term underlying mechanism of clinical activity of imatinib, nilotinib efficacy and superior tolerability over interferon (IFN)-based and dasatinib in the treatment of CML. As the first-generation therapy.4,5 Imatinib is widely accepted as the standard therapy TKI in this class of agents, imatinib inhibits BCR-ABL, albeit to a for newly diagnosed chronic-phase CML patients in addition to lesser extent than nilotinib or dasatinib. Imatinib also inhibits all CML patients who have failed stem cell transplant or IFN-a c-KIT and platelet-derived growth factor receptor (PDGFR) at therapy.6,7 Imatinib serves as a model for the utility of targeted clinically relevant concentrations (Table 1). Nilotinib inhibits agents in the treatment of cancer. For a relatively few CML most imatinib-resistant BCR-ABL forms, except T315I, in vitro at patients, resistance to imatinib is present before, or develops 17 concentrations approved for and tested in patients. Nilotinib is 30-fold more potent than imatinib at inhibiting BCR-ABL Correspondence: Dr FJ Giles, CTRC at The UT Health Science Center (Table 2).18,19 Early data on nilotinib in newly diagnosed CML at San Antonio, Institute for Drug Development, 7979 Wurzbach patients suggest clinical activity with the potential to exceed Road, San Antonio, TX 78229, USA. 20 E-mail: [email protected] first-line imatinib efficacy, pending longer follow-up data. 18,19 Received 19 March 2009; revised 6 April 2009; accepted 17 April Similar to imatinib, nilotinib inhibits c-KIT (IC50 ¼ 90 nM) 13,19 2009; published online 28 May 2009 and PDGFR (IC50 ¼ 72 nM). Nilotinib also inhibits the Class effects of TKIs in the treatment of CML FJ Giles et al 1699 ABL-related kinase ARG, DDR1 kinase, the oxidoreductase 325-fold more potency than imatinib at inhibiting BCR-ABL NQO2,20,21 and has some activity against the ephrin receptor (Table 2).14 Dasatinib also inhibits c-KIT and PDGFR with EPHB4, but displays no activity against the Src family of greater potency, and has a broad range of inhibitory activity kinases.13,22 Similar to nilotinib, dasatinib also inhibits most against a number of other tyrosine kinases (Table 2), including imatinib-resistant BCR-ABL forms, except T315I, in vitro.14,23 the ephrin receptor tyrosine kinase, Src and Src family kinases However, it displays greater inhibition, with approximately (FGR, FYN, HCK, LCK, LYN and YES).22,24 Lack of selectivity in binding Src family kinases probably derives from the fact that, unlike imatinib and nilotinib, dasatinib binds to the active conformation of the ABL kinase domain. In its active conforma- tion, ABL bears remarkable structural resemblance to the Src family kinases.25 Although both nilotinib and dasatinib have shown activity against a range of BCR-ABL kinase mutations, there exist a number of specific mutations that are less sensitive to each of these agents. For nilotinib, these mutations include E255K/V, Y253H, F359V and T315I, and for dasatinib, these mutations include Q252H, E255K/V, F317L, V299L and T315I.26–28 The relationship of kinase inhibition profile with safety Wild-type ABL inhibition: cardiac events All currently approved TKIs for CML treatment inhibit wild-type ABL kinase. ABL is a key mediator of normal cardiac function and is expressed in the cardiac and other tissues (Figure 2). A link between the imatinib inhibition of ABL and cardiac events was first suggested by Kerkela et al.29 In this retrospective study, 10 subjects with normal baseline cardiac function developed left ventricular dysfunction during imatinib therapy. Correlative studies in mice and cell lines revealed that imatinib-induced inhibition of ABL led to an endoplasmic reticulum stress response, collapse of the mitochondrial membrane potential, Table 2 IC50 values for bosutinib, imatinib, dasatinib and nilotinib against BCR-ABL expressed in Ba/F3-transfected cells108 IC50 values (nM) Bosutinib Imatinib Dasatinib Nilotinib BCR-ABL 41.61 527 1.83 17.69 Figure 1 Structures of current and investigational tyrosine kinase inhibitors for the treatment of CML.100 Adapted from Weisberg et al.106 Adapted from Redaelli et al.108 Table 1 Novel tyrosine kinase inhibitors currently in clinical trials for the treatment of CML105 SPOTLIGHT Tyrosine kinase inhibitor Kinase targets Drug Drug Company administration development status Imatinib (STI571) ABL, PDGFR, c-KIT Oral Approved Novartis Pharmaceuticals, San Carlos, CA, USA Dasatinib (BMS-34825) ABL, PDGFR, c-KIT, FGR, FYN, HCK, LCK, Oral Approved Bristol-Myers Squibb, LYN, SRC, YES, EPHB4 New York, NY, USA Nilotinib (AMN 107) ABL, PDGFR, c-KIT, ARG, EPHB4 Oral Approved Novartis, San Carlos, CA, USA Bosutinib (SKI-606) ABL, FGR, LYN, SRC Oral Phase II Wyeth, Madison, NJ, USA INNO-406 (NS-187) ABL, LYN, PDGFR, c-KIT Oral Phase I Innovive Pharmaceuticals, New York, NY, USA AZD0530 Src family kinases Oral Phase II AstraZeneca, Washington, DC, (solid tumors) USA MK-0457 (VX-680) Aurora kinases, FLTE, JAK2, ABL, T315I ABL Intravenous Phase II Merck & Co., Inc., Whitehouse Station, NJ, USA PHA-739358 Aurora A, B and C Oral Phase II Nerviano Medical Sciences, Nerviano, Italy Abbreviation: CML, chronic myeloid leukemia. Adapted from Weisberg et al.106 Leukemia Class effects of TKIs in the treatment of CML FJ Giles et al 1700 SPOTLIGHT Figure 2 ABL signaling and inhibition in cardiomyocytes.101 In cardiomyocytes, imatinib induces ER stress, leading to the activation of the PKR- like ER kinase (PERK) and IRE1 pathways, and to the overexpression of protein kinase Cd (PKCd).
Recommended publications
  • Promoter Janus Kinase 3 Proximal Characterization and Analysis Of
    The Journal of Immunology Characterization and Analysis of the Proximal Janus Kinase 3 Promoter1 Martin Aringer,2*† Sigrun R. Hofmann,2* David M. Frucht,* Min Chen,* Michael Centola,* Akio Morinobu,* Roberta Visconti,* Daniel L. Kastner,* Josef S. Smolen,† and John J. O’Shea3* Janus kinase 3 (Jak3) is a nonreceptor tyrosine kinase essential for signaling via cytokine receptors that comprise the common ␥-chain (␥c), i.e., the receptors for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Jak3 is preferentially expressed in hemopoietic cells and is up-regulated upon cell differentiation and activation. Despite the importance of Jak3 in lymphoid development and immune function, the mechanisms that govern its expression have not been defined. To gain insight into this issue, we set out to characterize the Jak3 promoter. The 5؅-untranslated region of the Jak3 gene is interrupted by a 3515-bp intron. Upstream of this intron and the transcription initiation site, we identified an ϳ1-kb segment that exhibited lymphoid-specific promoter activity and was responsive to TCR signals. Truncation of this fragment revealed that core promoter activity resided in a 267-bp fragment that contains putative Sp-1, AP-1, Ets, Stat, and other binding sites. Mutation of the AP-1 sites significantly diminished, whereas mutation of the Ets sites abolished, the inducibility of the promoter construct. Chromatin immunoprecipitation assays showed that histone acetylation correlates with mRNA expression and that Ets-1/2 binds this region. Thus, transcription factors that bind these sites, especially Ets family members, are likely to be important regulators of Jak3 expression.
    [Show full text]
  • Platelet-Derived Growth Factor Receptor Expression and Amplification in Choroid Plexus Carcinomas
    Modern Pathology (2008) 21, 265–270 & 2008 USCAP, Inc All rights reserved 0893-3952/08 $30.00 www.modernpathology.org Platelet-derived growth factor receptor expression and amplification in choroid plexus carcinomas Nina N Nupponen1,*, Janna Paulsson2,*, Astrid Jeibmann3, Brigitte Wrede4, Minna Tanner5, Johannes EA Wolff 6, Werner Paulus3, Arne O¨ stman2 and Martin Hasselblatt3 1Molecular Cancer Biology Program, University of Helsinki, Helsinki, Finland; 2Department of Oncology–Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden; 3Institute of Neuropathology, University Hospital Mu¨nster, Mu¨nster, Germany; 4Department of Pediatric Oncology, University of Regensburg, Regensburg, Germany; 5Department of Oncology, Tampere University Hospital, Tampere, Finland and 6Children’s Cancer Hospital, MD Anderson Cancer Center, Houston, TX, USA Platelet-derived growth factor (PDGF) receptor signaling has been implicated in the development of glial tumors, but not yet been examined in choroid plexus carcinomas, pediatric tumors with dismal prognosis for which novel treatment options would be desirable. Therefore, protein expression of PDGF receptors a and b as well as amplification status of the respective genes, PDGFRA and PDGFRB, were examined in a series of 22 patients harboring choroid plexus carcinoma using immunohistochemistry and chromogenic in situ hybridization (CISH). The majority of choroid plexus carcinomas expressed PDGF receptors with 6 cases (27%) displaying high staining scores for PDGF receptor a and 13 cases (59%) showing high staining scores for PDGF receptor b. Correspondingly, copy-number gains of PDGFRA were observed in 8 cases out of 12 cases available for CISH and 1 case displayed amplification (six or more signals per nucleus). The proportion of choroid plexus carcinomas with amplification of PDGFRB was even higher (5/12 cases).
    [Show full text]
  • Overexpresssion of Tyrosine Kinase Discoidin Domain Receptor I (DDR1 ) in Transitional Cell Carcinoma
    Overexpresssion of Tyrosine Kinase Discoidin domain receptor I (DDR1 ) in Transitional cell Carcinoma Szu-Ting Chen,* and Shie-Liang Hsieh* Institute and Department of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan; To whom proofs are to be sent: Shie-Liang Hsieh, Department and Institute of Microbiology and Immunology, National Yang-Ming University, Shih-Pai, Taipei 112, Taiwan. E-mail address: [email protected] Telephone number: 886-2-28267161 Fax number: 886-2-28277933 INTRODUCTION DDR1, discoidin domain receptor 1, belongs to the novel subfamily of tyrosine kinase receptor, which forms homodimer upon ligand engagement. DDR1 is distinguished from other receptor tyrosine kinase by the discoidin domain in their extracellular domain, which is a homology region originally identified in Dictyostelium discoideum (slime mold) protein, discoidin I, and involves in cells aggregation. Discoidin-1 has binding specificity toward galactose and N-acetyl galactosamine and is essential for slime mold cells adhesion, migration and aggregation during its development, suggesting that DDR1 shared similar biologically function in mammalian [1-3]. DDR1 has been found mainly distributed and in human tissue epithelia, such as kidney, breast, lung [3], bronchial [4] and keratinocytes [5]. Furthermore, DDR1 has been also reported its expression in immune system like the monocyte-derived dendritic cells, annotated as CD167 [6] and macrophage[7]. However, recently, the overexpression of DDR1 has been detected in several human cancers, such as primary breast cancer [1, 8, 9] ovarian[10, 11], brain[12], esophageal cancer [13], and TCC (our data unpublished) in which raising the possibility that DDR1 may play a important role in tumorigenesis [14].
    [Show full text]
  • Human and Mouse CD Marker Handbook Human and Mouse CD Marker Key Markers - Human Key Markers - Mouse
    Welcome to More Choice CD Marker Handbook For more information, please visit: Human bdbiosciences.com/eu/go/humancdmarkers Mouse bdbiosciences.com/eu/go/mousecdmarkers Human and Mouse CD Marker Handbook Human and Mouse CD Marker Key Markers - Human Key Markers - Mouse CD3 CD3 CD (cluster of differentiation) molecules are cell surface markers T Cell CD4 CD4 useful for the identification and characterization of leukocytes. The CD CD8 CD8 nomenclature was developed and is maintained through the HLDA (Human Leukocyte Differentiation Antigens) workshop started in 1982. CD45R/B220 CD19 CD19 The goal is to provide standardization of monoclonal antibodies to B Cell CD20 CD22 (B cell activation marker) human antigens across laboratories. To characterize or “workshop” the antibodies, multiple laboratories carry out blind analyses of antibodies. These results independently validate antibody specificity. CD11c CD11c Dendritic Cell CD123 CD123 While the CD nomenclature has been developed for use with human antigens, it is applied to corresponding mouse antigens as well as antigens from other species. However, the mouse and other species NK Cell CD56 CD335 (NKp46) antibodies are not tested by HLDA. Human CD markers were reviewed by the HLDA. New CD markers Stem Cell/ CD34 CD34 were established at the HLDA9 meeting held in Barcelona in 2010. For Precursor hematopoetic stem cell only hematopoetic stem cell only additional information and CD markers please visit www.hcdm.org. Macrophage/ CD14 CD11b/ Mac-1 Monocyte CD33 Ly-71 (F4/80) CD66b Granulocyte CD66b Gr-1/Ly6G Ly6C CD41 CD41 CD61 (Integrin b3) CD61 Platelet CD9 CD62 CD62P (activated platelets) CD235a CD235a Erythrocyte Ter-119 CD146 MECA-32 CD106 CD146 Endothelial Cell CD31 CD62E (activated endothelial cells) Epithelial Cell CD236 CD326 (EPCAM1) For Research Use Only.
    [Show full text]
  • 2025.Full-Text.Pdf
    Human Cancer Biology Inhibition of Phosphotyrosine Phosphatase 1B Causes Resistance in BCR-ABL-PositiveLeukemiaCellstotheABLKinaseInhibitorSTI571 Noriko Koyama,1Steffen Koschmieder,1SandhyaTyagi,1Ignacio Portero-Robles,1Jo« rg Chromic,1 Silke Myloch,1Heike Nu« rnberger,1Ta nja Ro s s m a ni t h , 1Wolf-Karsten Hofmann,2 Dieter Hoelzer,1and Oliver Gerhard Ottmann1 Abstract Protein tyrosine phosphatase 1B(PTP1B) is a negative regulator of BCR-ABL-mediated transfor- mation in vitro and in vivo. Toinvestigate whether PTP1B modulates the biological effects of the abl kinase inhibitor STI571in BCR-ABL-positive cells, we transfected Philadelphia chromosome ^ positive (Ph+) chronic myeloid leukemia cell-derived K562 cells with either wild-type PTP1B (K562/PTP1B), a substrate-trapping dominant-negative mutant PTP1B(K562/D181A), or empty vector (K562/mock). Cells were cultured with or without STI571and analyzed for its effects on proliferation, differentiation, and apoptosis. In both K562/mock and K562/PTP1B cells, 0.25 to 1 Amol/L STI571 induced dose-dependent growth arrest and apoptosis, as measured by a decrease of cell proliferation and an increase of Annexin V-positive cells and/or of cells in the sub-G1 apoptotic phase. Western blot analysis showed increased protein levels of activated caspase-3 and caspase-8 and induction of poly(ADP-ribose) polymerase cleavage. Low con- centrations of STI571promoted erythroid differentiation of these cells. Conversely, K562/D181A cells displayed significantly lower PTP1B-specific tyrosine phosphatase activity and were signifi- cantly less sensitive to STI571-induced growth arrest, apoptosis, and erythroid differentiation. Pharmacologic inhibition of PTP1B activity in wild-type K562 cells, using bis(N,N-dimethylhy- droxamido)hydroxooxovanadate, attenuated STI571-induced apoptosis.
    [Show full text]
  • Bosutinib (Bosulif®) (“Boe SUE Ti Nib”)
    Bosutinib (Bosulif®) (“boe SUE ti nib”) How drug is given: By mouth Purpose: To stop the growth of cancer cells in chronic myelogenous leukemia (CML). How to take this drug 1. Take this medication with food. 2. Swallow each tablet whole; do not crush or chew. IF you have trouble swallowing the tablet, the pharmacist will give you specific instructions. 3. IF you miss a dose, take it as soon as possible. However, if it is less than 12 hours until your next dose, skip the missed dose and go back to your regular dosing schedule. Do not double dose. 4. Wash hands after taking the medication. Avoid handling crushed or broken tablets. 5. Do not drink grapefruit juice or eat grapefruit. Also, do not take St. John’s wort. Consuming these may increase the amount of medication in your body and worsen side effects. 6. Bosutinib can interFere with many drugs, which may change how this works in your body. Talk with your doctor before starting any new drugs, including over the counter drugs, natural products, herbals or vitamins. This includes drugs such as Prilosec™. Storage • Store this medication at room temperature 68-77°F (20-25°C), away From heat, moisture, and direct light. Keep this medicine in its original container, out oF reach oF children and pets. Things that may occur during treatment 1. Loose stools or diarrhea may occur within a few days after the drug is started. You may take loperamide (Imodium A-D®) to help control diarrhea. You may buy this at most drug stores.
    [Show full text]
  • CD137 Microbead Kit CD137 Microbead + Cells
    CD137 MicroBead Kit human Order no. 130-093-476 Contents 1.2 Background information 1. Description The activation-induced antigen CD137 (4-1BB) is a 30 kDa glycoprotein of the tumor necrosis factor (TNF) receptor 1.1 Principle of the MACS® Separation + + superfamily. It is mainly expressed on activated CD4 and CD8 1.2 Background information T cells, activated B cells, and natural killer cells, but can also be 1.3 Applications found on resting monocytes and dendritic cells. As a costimulatory molecule, CD137 is involved in the activation 1.4 Reagent and instrument requirements and survival of CD4, CD8, and NK cells. Its engagement enhances 2. Protocol expansion of T cells and activates them to secrete cytokines. CD137 has been described to be a suitable marker for antigen- 2.1 Sample preparation specific activation of human CD8+ T cells, as CD137 is not expressed 2.2 Magnetic labeling on resting CD8+ T cells and its expression is reliably induced after 2.3 Magnetic separation 24 hours of stimulation.¹,² 3. Example of a separation using the CD137 MicroBead Kit 1.3 Applications 4. References ● Enrichment of CD137+ T cells for phenotypical and functional 5. Appendix characterization. ● Enrichment of activated antigen-specific T cells after antigen- Warnings specific stimulation. Reagents contain sodium azide. Under acidic conditions sodium 1.4 Reagent and instrument requirements azide yields hydrazoic acid, which is extremely toxic. Azide ● compounds should be diluted with running water before discarding. Buffer: Prepare a solution containing phosphate-buffered These precautions are recommended to avoid deposits in plumbing saline (PBS), pH 7.2, 0.5% bovine serum albumin (BSA), where explosive conditions may develop.
    [Show full text]
  • Insulin Receptor Substrates-5 and -6 Are Poor Substrates for the Insulin Receptor
    MOLECULAR MEDICINE REPORTS 3: 189-193, 2010 189 Insulin receptor substrates-5 and -6 are poor substrates for the insulin receptor SoETkIn VERSTEyHE1, CHRISToPHE BlanqUart2,3, CoRnElIa HamPE2,3, SHaUkaT maHmooD1, nEVEna CHRISTEFF2,3, PIERRE DE MEYTS1, STEVEn G. GRay1,4 and TARIK ISSAD2,3 1Receptor Systems Biology laboratory, Hagedorn Research Institute, novo nordisk a/S, 2820 Gentofte, Denmark; 2Institut Cochin, Université Paris Descartes, CnRS (UmR 8104), 75014 Paris; 3Inserm, U567, 75654 Paris, France; 4Translational Cancer Research Group, Trinity Centre for Health Sciences, Institute of molecular medicine, St James's Hospital, Dublin 8, Ireland Received September 30, 2009; accepted november 13, 2009 DoI: 10.3892/mmr_00000239 Abstract. Insulin receptor substrates (IRS)-5 and -6 are two leads to the activation of receptor tyrosine kinase and receptor recently identified members of the IRS family. We investi- phosphorylation, which enables the binding of docking proteins gated their roles as insulin receptor substrates and compared such as insulin receptor substrates (IRS)-1, -2, -3 and -4 and them with Src-homology-2-containing (Shc) protein, a Src-homology-2-containing protein (Shc). This in turn leads to well-established substrate. Bioluminescence resonance their phosphorylation and, thereby, intracellular signalling (1). energy transfer (BRET) experiments showed no interaction Until recently, the IRS family included IRS-1, -2, -3 and between the receptor and IRS-5, while interaction with IRS-6 -4 (2) and three downstream of kinases, Dok-1, -2 and -3. was not enhanced by insulin. By contrast, Shc showed an These seven proteins have similar amino-terminal pleckstrin insulin-induced BRET response, as did a truncated form of homology (PH) and similar phosphotyrosine binding (PTB) IRS-1 (1-262).
    [Show full text]
  • Paxillin Binding to the Cytoplasmic Domain of CD103 Promotes Cell Adhesion and Effector
    Author Manuscript Published OnlineFirst on October 11, 2017; DOI: 10.1158/0008-5472.CAN-17-1487 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Paxillin binding to the cytoplasmic domain of CD103 promotes cell adhesion and effector functions for CD8+ resident memory T cells in tumors Ludiane Gauthier1, Stéphanie Corgnac1, Marie Boutet1, Gwendoline Gros1, Pierre Validire2, Georges Bismuth3 and Fathia Mami-Chouaib1 1 INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France 2 Institut Mutualiste Montsouris, Service d’Anatomie pathologique, 75014 Paris, France. 3 INSERM U1016, CNRS UMR8104, Université Paris Descartes, Institut Cochin, 75014 Paris. S Corgnac, M Boutet and G Gros contributed equally to this work. M Boutet current address: Department of Microbiology and Immunology Albert Einstein College of Medecine, NY 10461 USA. Corresponding author: Fathia Mami-Chouaib, INSERM UMR 1186, Gustave Roussy. 39, rue Camille Desmoulins, F-94805 Villejuif. Phone: +33 1 42 11 49 65, Fax: +33 1 42 11 52 88, e-mail: [email protected] and [email protected] Running title: CD103 signaling in human TRM cells Key words: TRM cells, CD103 integrin, T-cell function and signaling, paxillin. Abbreviations: IS: immune synapse; LFA: leukocyte function-associated antigen; FI: fluorescence intensity; mAb: monoclonal antibody; phospho: phosphorylated; Pyk2: proline- rich tyrosine kinase-2; NSCLC: non-small-cell lung carcinoma; r: recombinant; sh-pxn: shorthairpin RNA-paxillin; TCR: T-cell receptor; TIL: tumor-infiltrating lymphocyte; TRM: tissue-resident memory T.
    [Show full text]
  • Calycosin Down-Regulates C-Met to Suppress Development of Glioblastomas
    J Biosci (2019) 44:96 Ó Indian Academy of Sciences DOI: 10.1007/s12038-019-9904-4 (0123456789().,-volV)(0123456789().,-volV) Calycosin down-regulates c-Met to suppress development of glioblastomas , XIAOHU NIE ,YUE ZHOU* ,XIAOBING LI,JIE XU,XUYAN PAN,RUI YIN and BIN LU Department of Neurosurgery, Huzhou Central Hospital, Huzhou, Zhejiang, People’s Republic of China *Corresponding author (Email, [email protected]) These authors contributed equally to this work. MS received 15 October 2018; accepted 9 June 2019; published online 7 August 2019 The antitumor effect of calycosin has been widely studied, but the targets of calycosin against glioblastomas are still unclear. In this study we focused on revealing c-Met as a potential target of calycosin suppressing glioblastomas. In this study, suppressed-cell proliferation and cell invasion together with induced-cell apoptosis appeared in calycosin-treated U251 and U87 cells. Under treatment of calycosin, the mRNA expression levels of Dtk, c-Met, Lyn and PYK2 were observed in U87 cells. Meanwhile a western blot assay showed that c-Met together with matrix metalloproteinases-9 (MMP9) and phosphorylation of the serine/threonine kinase AKT (p-AKT) was significantly down-regulated by calycosin. Furthermore, overexpressed c-Met in U87 enhanced the expression level of MMP9 and p-AKT and also improved cell invasion. Additionally, the expression levels of c-Met, MMP9 and p-AKT were inhibited by calycosin in c-Met overex- pressed cells. However, an AKT inhibitor (LY294002) only effected on MMP9 and p-AKT, not on c-Met. These data collectively indicated that calycosin possibility targeting on c-Met and exert an anti-tumor role via MMP9 and AKT.
    [Show full text]
  • Insulin Activates the Insulin Receptor to Downregulate the PTEN Tumour Suppressor
    Oncogene (2014) 33, 3878–3885 OPEN & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc ORIGINAL ARTICLE Insulin activates the insulin receptor to downregulate the PTEN tumour suppressor J Liu1, S Visser-Grieve2,4, J Boudreau1, B Yeung2,SLo1, G Chamberlain1,FYu1, T Sun3,5, T Papanicolaou1, A Lam1, X Yang2 and I Chin-Sang1 Insulin and insulin-like growth factor-1 signaling have fundamental roles in energy metabolism, growth and development. Recent research suggests hyperactive insulin receptor (IR) and hyperinsulinemia are cancer risk factors. However, the mechanisms that account for the link between the hyperactive insulin signaling and cancer risk are not well understood. Here we show that an insulin-like signaling inhibits the DAF-18/(phosphatase and tensin homolog) PTEN tumour suppressor in Caenorhabditis elegans and that this regulation is conserved in human breast cancer cells. We show that inhibiting the IR increases PTEN protein levels, while increasing insulin signaling decreases PTEN protein levels. Our results show that the kinase region of IRb subunit physically binds to PTEN and phosphorylates on Y27 and Y174. Our genetic results also show that DAF-2/IR negatively regulates DAF-18/PTEN during C. elegans axon guidance. As PTEN is an important tumour suppressor, our results therefore suggest a possible mechanism for increased cancer risk observed in hyperinsulinemia and hyperactive IR individuals. Oncogene (2014) 33, 3878–3885; doi:10.1038/onc.2013.347; published online 2 September 2013 Keywords: PTEN; C. elegans; tumour suppressor; cancer; insulin signaling; genetics INTRODUCTION tumour suppression and further substantiate the importance of Insulin and insulin-like growth factor (IGF)-1 signaling are the PTEN regulation in cancer progression.
    [Show full text]
  • Paxillin: a Focal Adhesion-Associated Adaptor Protein
    Oncogene (2001) 20, 6459 ± 6472 ã 2001 Nature Publishing Group All rights reserved 0950 ± 9232/01 $15.00 www.nature.com/onc Paxillin: a focal adhesion-associated adaptor protein Michael D Schaller*,1 1Department of Cell and Developmental Biology, Lineberger Comprehensive Cancer Center and Comprehensive Center for In¯ammatory Disorders, University of North Carolina, Chapel Hill, North Carolina, NC 27599, USA Paxillin is a focal adhesion-associated, phosphotyrosine- The molecular cloning of paxillin revealed a number containing protein that may play a role in several of motifs that are now known to function in mediating signaling pathways. Paxillin contains a number of motifs protein ± protein interactions (see Figure 1) (Turner that mediate protein ± protein interactions, including LD and Miller, 1994; Salgia et al., 1995a). The N-terminal motifs, LIM domains, an SH3 domain-binding site and half of paxillin contains a proline-rich region that SH2 domain-binding sites. These motifs serve as docking could serve as an SH3 domain-binding site. Several sites for cytoskeletal proteins, tyrosine kinases, serine/ tyrosine residues conforming to SH2 domain binding threonine kinases, GTPase activating proteins and other sites were also noted. In addition, the N-terminal adaptor proteins that recruit additional enzymes into domain of paxillin contains ®ve copies of a peptide complex with paxillin. Thus paxillin itself serves as a sequence, called the LD motif, which are now known docking protein to recruit signaling molecules to a to function as binding sites for other proteins (see speci®c cellular compartment, the focal adhesions, and/ Table 1) (Brown et al., 1998a). The C-terminal half of or to recruit speci®c combinations of signaling molecules paxillin is comprised of four LIM domains, which are into a complex to coordinate downstream signaling.
    [Show full text]