Activity of Dasatinib, a Dual SRC/ABL Kinase Inhibitor, and IPI-504, A

Total Page:16

File Type:pdf, Size:1020Kb

Activity of Dasatinib, a Dual SRC/ABL Kinase Inhibitor, and IPI-504, A Cancer Therapy: Preclinical Activity of Dasatinib, a Dual SRC/ABL Kinase Inhibitor, and IPI-504, aHeatShockProtein90Inhibitor,againstGastrointestinal Stromal Tumor ^ Associated PDGFRAD842V Mutation Barbara Dewaele,1BartoszWasag,1Jan Cools,1,4 Raf Sciot,2 Hans Prenen,3 PeterVandenberghe,1 AgnieszkaWozniak,3 Patrick Scho« ffski,3 Peter Marynen,1,4 and Maria Debiec-Rychter1 Abstract Purpose: Activating mutations in platelet-derived growthfactor receptor- a (PDGFRA) have been reported in f5% to 10% of patients withgastrointestinal stromal tumors (GIST). Imatinib efficiently inhibits the juxtamembrane PDGFRA mutations, whereas many tyrosine kinase domain activation loop PDGFRA mutations confer primary resistance to imatinib. In this study, we compared the efficacy of second-line tyrosine kinase inhibitors such as dasatinib, sorafenib, and nilotinib against two GIST-related PDGFRA mutants, PDGFRAD842V and PDGFRADDIM842-844. In addition, we sought to investigate the inhibitory effect of the heat shock protein 90 inhibitor, IPI-504, on these mutants. Experimental Design: Primary imatinib-resistant tumor cells and cell lines expressing imatinib- resistant PDGFRAD842V or imatinib-sensitive PDGFRADDIM842-844 mutants were treated with different concentrations of dasatinib, sorafenib, nilotinib, and IPI-504. The effect of treatment on proliferation, survival, and signaling was determined. Results: All inhibitors tested exhibited a high efficacy toward the PDGFRADDIM842-844 mutant. In contrast, ex vivo and in vitro assays revealed that only dasatinib potently inhibited the D842V PDGFRA isoform withan IC 50 value of 62 nmol/L. Sorafenib and nilotinib were significantly less efficacious against this mutation, inhibiting the PDGFRA kinase activity at >1, 0 0 0 a nd >5,000 nmol/L, and suppressing the proliferation of the cells expressing the PDGFRAD842V mutant withan IC 50 value of 239 and 1,310 nmol/L, respectively. IPI-504 treatment potently inhibited PDGFRA kinase activity by inducing the degradation of PDGFRAD842V and PDGFRADDIM842-844 at 256 and 182 nmol/L, respectively. Conclusions: Treatment with dasatinib or the heat shock protein 90 inhibitor IPI-504 may provide a therapeutic alternative for GIST patients whose tumors carry the imatinib-resistant PDGFRAD842V mutant isoform. Gastrointestinal stromal tumors (GIST) comprise the largest tyrosine kinase homologous to KIT (5). Activating KIT and subset of mesenchymal tumors that develop along the PDGFRA mutations in GISTs differ in type and affect different gastrointestinal tract (1, 2). Constitutive activation of the KIT receptor domains (6). Therapeutic inhibition of KIT or tyrosine kinase through oncogenic mutations is an early and PDGFRA by the small-molecule inhibitor imatinib gives a probably initiating event in the majority of GISTs (3, 4). clinical response in the majority of advanced GISTs (3). Alternatively, a subset of GISTs express a constitutively activated However, early resistance has been reported in 10% to 20% platelet-derived growth factor receptor-a (PDGFRA) mutant, a of patients. It is well known that sensitivity to imatinib depends on the location of the KIT and PDGFRA gene mutation (3). Based on in vitro studies and subsequent clinical trials, it has been shown that a subset of GISTs harboring KIT and PDGFRA 1 2 3 Authors’Affiliations: Departments of Human Genetics, Pathology, and General mutations in the second tyrosine kinase domain (activation Medical Oncology, K.U. Leuven, and 4VIB Department of Molecular and Developmental Genetics,VIB, Leuven, Belgium loop domain) do not respond well to imatinib treatment Received 2/27/08; revised 4/28/08; accepted 5/22/08. (3, 7–10). The oncogenic PDGFRA mutations detected in Grant support: Life Raft Group, the Fonds voor Wetenschappelijk Onderzoek GISTs mainly involve exons 12, 14, or 18, and account for Vlaanderen (G.0286.05; M. Debiec-Rychter), and by a Concerted Action grant f5% to 10% of all GIST mutations. Wild-type PDGFRA is 2006/14 from the K.U. Leuven. sensitive to imatinib (11), but only a portion of the consti- The costs of publication of this article were defrayed in part by the payment of page advertisement tutively activated PDGFRA isoforms identified in GISTs are charges. This article must therefore be hereby marked in accordance D842V with18 U.S.C. Section 1734 solely to indicate this fact. potently inhibited by imatinib (3). The missense PDGFRA Requests for reprints: Maria Debiec-Rychter, Department of Human Genetics, mutation resulting in the substitution of aspartic acid to valine University of Leuven, O&N Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium. at codon 842 accounts for f60% of all PDGFRA mutations Phone: 32-16-347218; Fax: 32-16-346210; E-mail: Maria.Debiec-Rychter@ med.kuleuven.be. known in GISTs (5). It confers primary resistance to imatinib F 2008 American Association for Cancer Research. in vitro and in vivo (3, 5, 8, 12), resistance to sunitinib in vitro doi:10.1158/1078-0432.CCR-08-0533 (13), and resistance to nilotinib in vitro and in vivo (14). www.aacrjournals.org 5749 Clin Cancer Res 2008;14(18) September 15, 2008 Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2008 American Association for Cancer Research. Cancer Therapy: Preclinical Conversely, we have previously shown that PKC412 efficiently oncoprotein degradation, but the effect of HSP90 inhibitors on inhibited the PDGFRAD842V mutant at the concentration of GISTs bearing PDGFRA mutations has thus far not been tested. 1 Amol/L (12). In this report, we examined the efficacy of second-generation Because resistance of GIST patients to imatinib is a tyrosine kinase inhibitors nilotinib, dasatinib, and sorafenib for D continuous clinical challenge, multiple novel therapeutic the inhibition of PDGFRAD842V-expressing and PDGFRA DIM842-844– strategies are under development. Novel small molecule expressing cells. In addition, we tested the potency of the HSP90 compounds being tested in clinical trials include nilotinib, inhibitor, IPI-504, to induce PDGFRA oncoprotein degradation sorafenib, and dasatinib. as a therapeutic alternative for imatinib-resistant, PDGFRA mutant In addition, heat shock protein 90 (HSP90) inhibition, GISTs. causing degradation of wild-type and imatinib-resistant KIT mutant proteins, has recently been validated in preclinical Materials and Methods studies as an alternative therapeutic strategy to challenge the heterogeneity of KIT imatinib-resistant mutations in patients Inhibitors. The inhibitors, imatinib mesylate (Glivec/Gleevec, with GIST (15). The PDGFRA protein is also subject to HSP90- Novartis), sorafenib (BAY-43-9006, Bayer AG), nilotinib (AMN107, mediated protection from proteasomal degradation in cancer Novartis), dasatinib (BMS-354825, Bristol-Meyers-Squibb), and IPI-504 cells (16) and HSP90 inhibition may cause effective PDGFRA were provided by Infinity Pharmaceuticals, Inc. and MedImmune Inc. Fig. 1. In vitro effect of imatinib, nilotinib, sorafenib, and dasatinib on PDGFRADDIM842-844 ^expressingandPDGFRAD842V-expressing Ba/F3 cells. The dose-response proliferation curves of PDGFRADDIM842-844 ^ transduced (A)andPDGFRAD842V-transduced (B) Ba/F3 cells, treated withimatinib, nilotinib, sorafenib, and dasatinib for 48 h. Points, the average results of two experiments done in triplicate plotted with the curve-fitting Origin software; bars, SD.The calculated IC50 for eachinhibitor is indicated. Representative results from apoptotic assays of PDGFRADDIM842-844 ^ transduced (C)andPDGFRAD842V-transduced (D) Ba/F3 cells. All compounds induced significant apoptosisbetween100and500nmol/LincellsexpressingPDGFRADDIM842-844. In contrast, only dasatinib showed significant induction of apoptosis at 500 nmol/L in cells expressing PDGFRAD842V. Clin Cancer Res 2008;14(18) September 15, 2008 5750 www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2008 American Association for Cancer Research. Therapeutic Alternative for PDGFRAD842V Mutant Table 1. Comparison of in vitro apoptosis induction of imatinib-sensitive PDGFRADDIM842-844 and imatinib- resistant PDGFRAD842V mutants expressed in the Ba/F3 cells PDGFRA#DIM842-844 PDGFRAD842V Normal (%) Necrotic (%) Apoptotic (%) Normal (%) Necrotic (%) Apoptotic (%) Vehicle 85.1 9 5.7 84.2 7.6 7.4 Imatinib 0.1 Amol/L 71.8 15.1 12.9 85.9 6.8 6.6 0.5 Amol/L 60.9 20.8 18.1 87.7 4.7 7.3 Nilotinib 0.1 Amol/L 85 7.8 7.1 87.7 6.9 4.5 0.5 Amol/L 73.8 13.4 12.5 88.4 5.7 5.2 Sorafenib 0.1 Amol/L 74.1 14.1 11.5 87.9 6.6 5 0.5 Amol/L 66.5 16.7 16.8 89.4 5.9 4.5 Dasatinib 0.1 Amol/L 46.1 26.6 27 77.9 13.3 8.3 0.5 Amol/L 39.3 30.3 28.9 59.4 24.1 15.4 IPI-504 0.1 Amol/L 73.1 14.6 12.1 80.3 11.9 6.5 0.5 Amol/L 48.4 28.6 22.1 37.8 37.8 22.8 A 10 mmol/L stock solution of the inhibitors, dissolved in DMSO or a citric salt buffer for IPI-504) and incubated for 2 h at +37jC. After a citric salt buffer (for IPI-504) was stored at -80jC. The inhibitors were wash in ice-cold PBS, cells were lysed. Lysates were separated by SDS- diluted in culture medium just before use. PAGE electrophoresis and immunoblotted using anti–phospho- In vitro assays using transduced Ba/F3 cells. The retroviral expres- PDGFRA(Tyr754), anti-PDGFRA, and anti-actin antibodies. D sion constructs pMSCV-PDGFRAD842V and pMSCV-PDGFRA DIM842-844 The 5-bromo-2¶-deoxyuridine cell proliferation (Roche Applied were described previously (12). Retrovirally transduced Ba/F3 cells, Science) and sulforhodamine B survival (Sigma-Aldrich) assays were stably expressing the above PDGFRA constructs, were maintained in used to evaluate the proliferation rate and the survival of primary GIST culture at 1 Â 106 cells/mL. For the Ba/F3 dose-response assays, cells, according to the manufacturer’s instructions (18, 19). Both tests retrovirally transduced Ba/F3 cells were grown in RPMI 1640 supple- were run simultaneously and in duplicate. In short, the adherent mented with 10% fetal bovine serum, with or without interleukin 3, primary GIST cells were seeded in 96-well plates (10,000 cells per well) in the presence of vehicle alone or with varying concentrations of the and incubated overnight at +37jC.
Recommended publications
  • Management of Chronic Myelogenous Leukemia in Pregnancy
    ANTICANCER RESEARCH 35: 1-12 (2015) Review Management of Chronic Myelogenous Leukemia in Pregnancy AMIT BHANDARI, KATRINA ROLEN and BINAY KUMAR SHAH Cancer Center and Blood Institute, St. Joseph Regional Medical Center, Lewiston, ID, U.S.A. Abstract. Discovery of tyrosine kinase inhibitors has led to Leukemia in pregnancy is a rare condition, with an annual improvement in survival of chronic myelogenous leukemia incidence of 1-2/100,000 pregnancies (8). Since the first (CML) patients. Many young CML patients encounter administration of imatinib (the first of the TKIs) to patients pregnancy during their lifetime. Tyrosine kinase inhibitors with CML in June 1998, it is estimated that there have now inhibit several proteins that are known to have important been 250,000 patient years of exposure to the drug (mostly functions in gonadal development, implantation and fetal in patients with CML) (9). TKIs not only target BCR-ABL development, thus increasing the risk of embryo toxicities. tyrosine kinase but also c-kit, platelet derived growth factors Studies have shown imatinib to be embryotoxic in animals with receptors α and β (PDGFR-α/β), ARG and c-FMS (10). varying effects in fertility. Since pregnancy is rare in CML, Several of these proteins are known to have functions that there are no randomized controlled trials to address the may be important in gonadal development, implantation and optimal management of this condition. However, there are fetal development (11-15). Despite this fact, there is still only several case reports and case series on CML in pregnancy. At limited information on the effects of imatinib on fertility the present time, there is no consensus on how to manage and/or pregnancy.
    [Show full text]
  • The Effects of Combination Treatments on Drug Resistance in Chronic Myeloid Leukaemia: an Evaluation of the Tyrosine Kinase Inhibitors Axitinib and Asciminib H
    Lindström and Friedman BMC Cancer (2020) 20:397 https://doi.org/10.1186/s12885-020-06782-9 RESEARCH ARTICLE Open Access The effects of combination treatments on drug resistance in chronic myeloid leukaemia: an evaluation of the tyrosine kinase inhibitors axitinib and asciminib H. Jonathan G. Lindström and Ran Friedman* Abstract Background: Chronic myeloid leukaemia is in principle a treatable malignancy but drug resistance is lowering survival. Recent drug discoveries have opened up new options for drug combinations, which is a concept used in other areas for preventing drug resistance. Two of these are (I) Axitinib, which inhibits the T315I mutation of BCR-ABL1, a main source of drug resistance, and (II) Asciminib, which has been developed as an allosteric BCR-ABL1 inhibitor, targeting an entirely different binding site, and as such does not compete for binding with other drugs. These drugs offer new treatment options. Methods: We measured the proliferation of KCL-22 cells exposed to imatinib–dasatinib, imatinib–asciminib and dasatinib–asciminib combinations and calculated combination index graphs for each case. Moreover, using the median–effect equation we calculated how much axitinib can reduce the growth advantage of T315I mutant clones in combination with available drugs. In addition, we calculated how much the total drug burden could be reduced by combinations using asciminib and other drugs, and evaluated which mutations such combinations might be sensitive to. Results: Asciminib had synergistic interactions with imatinib or dasatinib in KCL-22 cells at high degrees of inhibition. Interestingly, some antagonism between asciminib and the other drugs was present at lower degrees on inhibition.
    [Show full text]
  • Combinatorial Efficacy of Entospletinib and Chemotherapy in Patient-Derived Xenograft Models of Infant Acute Lymphoblastic Leukemia
    Acute Lymphoblastic Leukemia SUPPLEMENTARY APPENDIX Combinatorial efficacy of entospletinib and chemotherapy in patient-derived xenograft models of infant acute lymphoblastic leukemia Joseph P. Loftus, 1* Anella Yahiaoui, 2* Patrick A Brown, 3 Lisa M. Niswander, 1 Asen Bagashev, 1 Min Wang, 2 Allyson Shauf, 2 Stacey Tannheimer 2 and Sarah K. Tasian 1,4 1Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA; 2Gilead Sci - ences, Foster City, CA; 3Department of Pediatrics, Division of Pediatric Hematology/Oncology, Johns Hopkins University and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD and 4Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA *JPL and AY contributed equally as co-first authors. ©2021 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol. 2019.241729 Received: October 28, 2019. Accepted: May 8, 2020. Pre-published: May 15, 2020. Correspondence: SARAH K. TASIAN - [email protected] SUPPLEMENTAL DATA SUPPLEMENTAL METHODS Cell Titer Glo viability assays The human B-acute lymphoblastic leukemia (B-ALL) cell lines NALM-6 (non-KMT2A- rearranged) and HB11;19, KOPN-8, and HB11;19 (all KMT2A-rearranged) were obtained from the German Collection of Microorganisms and Cell Cultures GmbH (Deutsche Sammlung von Mikroorganismen und Zellkulturen; DSMZ), validated by short tandem repeat identity testing, and confirmed to be Mycoplasma-free. ALL cells were incubated in vitro with dimethyl sulfoxide or increasing concentrations of entospletinib (Gilead Sciences), fostamatinib (Selleckchem), dasatinib (LC Labs), selumetinib (LC Labs), and/or dexamethasone as indicated in Supplemental Figure 1 for 72 hours, then assessed for cell viability via Cell Titer Glo luminescent assays (Promega) according to manufacturer’s instructions using an IVIS Spectrum bioluminescent imaging instrument and Living Image software (PerkinElmer) (1, 2).
    [Show full text]
  • Antibody–Drug Conjugates: the Last Decade
    pharmaceuticals Review Antibody–Drug Conjugates: The Last Decade Nicolas Joubert 1,* , Alain Beck 2 , Charles Dumontet 3,4 and Caroline Denevault-Sabourin 1 1 GICC EA7501, Equipe IMT, Université de Tours, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France; [email protected] 2 Institut de Recherche Pierre Fabre, Centre d’Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160 Saint Julien en Genevois, France; [email protected] 3 Cancer Research Center of Lyon (CRCL), INSERM, 1052/CNRS 5286/UCBL, 69000 Lyon, France; [email protected] 4 Hospices Civils de Lyon, 69000 Lyon, France * Correspondence: [email protected] Received: 17 August 2020; Accepted: 10 September 2020; Published: 14 September 2020 Abstract: An armed antibody (antibody–drug conjugate or ADC) is a vectorized chemotherapy, which results from the grafting of a cytotoxic agent onto a monoclonal antibody via a judiciously constructed spacer arm. ADCs have made considerable progress in 10 years. While in 2009 only gemtuzumab ozogamicin (Mylotarg®) was used clinically, in 2020, 9 Food and Drug Administration (FDA)-approved ADCs are available, and more than 80 others are in active clinical studies. This review will focus on FDA-approved and late-stage ADCs, their limitations including their toxicity and associated resistance mechanisms, as well as new emerging strategies to address these issues and attempt to widen their therapeutic window. Finally, we will discuss their combination with conventional chemotherapy or checkpoint inhibitors, and their design for applications beyond oncology, to make ADCs the magic bullet that Paul Ehrlich dreamed of. Keywords: antibody–drug conjugate; ADC; bioconjugation; linker; payload; cancer; resistance; combination therapies 1.
    [Show full text]
  • 2021 Formulary List of Covered Prescription Drugs
    2021 Formulary List of covered prescription drugs This drug list applies to all Individual HMO products and the following Small Group HMO products: Sharp Platinum 90 Performance HMO, Sharp Platinum 90 Performance HMO AI-AN, Sharp Platinum 90 Premier HMO, Sharp Platinum 90 Premier HMO AI-AN, Sharp Gold 80 Performance HMO, Sharp Gold 80 Performance HMO AI-AN, Sharp Gold 80 Premier HMO, Sharp Gold 80 Premier HMO AI-AN, Sharp Silver 70 Performance HMO, Sharp Silver 70 Performance HMO AI-AN, Sharp Silver 70 Premier HMO, Sharp Silver 70 Premier HMO AI-AN, Sharp Silver 73 Performance HMO, Sharp Silver 73 Premier HMO, Sharp Silver 87 Performance HMO, Sharp Silver 87 Premier HMO, Sharp Silver 94 Performance HMO, Sharp Silver 94 Premier HMO, Sharp Bronze 60 Performance HMO, Sharp Bronze 60 Performance HMO AI-AN, Sharp Bronze 60 Premier HDHP HMO, Sharp Bronze 60 Premier HDHP HMO AI-AN, Sharp Minimum Coverage Performance HMO, Sharp $0 Cost Share Performance HMO AI-AN, Sharp $0 Cost Share Premier HMO AI-AN, Sharp Silver 70 Off Exchange Performance HMO, Sharp Silver 70 Off Exchange Premier HMO, Sharp Performance Platinum 90 HMO 0/15 + Child Dental, Sharp Premier Platinum 90 HMO 0/20 + Child Dental, Sharp Performance Gold 80 HMO 350 /25 + Child Dental, Sharp Premier Gold 80 HMO 250/35 + Child Dental, Sharp Performance Silver 70 HMO 2250/50 + Child Dental, Sharp Premier Silver 70 HMO 2250/55 + Child Dental, Sharp Premier Silver 70 HDHP HMO 2500/20% + Child Dental, Sharp Performance Bronze 60 HMO 6300/65 + Child Dental, Sharp Premier Bronze 60 HDHP HMO
    [Show full text]
  • Hypopigmentation of the Skin and Hair Associated with Targetedclinical Therapies and BIOMEDICAL INVESTIGATION
    M.T.J. Vega González, et al.: Hypopigmentation of the Skin and Hair Associated with TargetedCLINICAL Therapies AND BIOMEDICAL INVESTIGATION. 2014;1 REVIEW ARTICLE Journal of Cancerology. 2014;1:67-72 Hypopigmentation of the Skin and Hair Associated with Targeted Therapies MARÍA TERESA DE JESÚS VEGA GONZÁLEZ1, JORGE LUIS MARTÍNEZ TLAHUEL2 AND ELENA CINTHLELY MARTÍNEZ GUERRA3 1Skin and Soft Tissue Unit; 2Department of Medical Oncology. National Cancer Institute, Mexico City, Mexico; 3Dr. Ladislao de la Pascua Dermatology Center, Department of Health, Mexico City, Mexico © Permanyer Publications 2014 ABSTRACT In the last two decades the US Food and Drug Administration has granted approval for multiple molecules for use in the treatment of cancer. These targeted therapies, aimed at inhibiting specific molecules, cause numerous adverse effects at the skin, for example, acneform dermatitis, mucosal, hair and nail alterations, and depigmentation of skin and hair, which have been seldom described in the literature. In this article we will focus on the side effects of tyrosine kinase inhibitors in particular and try to show how these drugs affect melanogenesis in hair follicles, triggering changes in skin and hair pigmentation. (J CANCEROL. 2014;1:67-72) Corresponding author: Elena Cinthlely Martínez Guerra, [email protected] Key words: Depigmentation. Targeted therapy. Tyrosine kinase inhibitor. Correspondence to: Elena Cinthlely Martínez Guerra Dr. Vértiz, 464. Esq. Eje 3 Sur Col. Buenos Aires, Del. Cuauhtémoc 06780 México, D.F., México Received for publication: 31-10-2014 E-mail: [email protected] Accepted for publication: 18-11-2014 of the publisher. No part of this publication may be reproduced or photocopying without the prior written permission 67 Journal of Cancerology.
    [Show full text]
  • The Mitogen-Activated Protein Kinase Pathway Facilitates Resistance to the Src Inhibitor Dasatinib in Thyroid Cancer Thomas C
    Published OnlineFirst May 24, 2016; DOI: 10.1158/1535-7163.MCT-15-0702 Cancer Biology and Signal Transduction Molecular Cancer Therapeutics The Mitogen-Activated Protein Kinase Pathway Facilitates Resistance to the Src Inhibitor Dasatinib in Thyroid Cancer Thomas C. Beadnell1, Katie M. Mishall1, Qiong Zhou1, Stephen M. Riffert1, Kelsey E. Wuensch1, Brittelle E. Kessler1, Maia L. Corpuz1, Xia Jing1, Jihye Kim2, Guoliang Wang2, Aik Choon Tan2,3, and Rebecca E. Schweppe1,3 Abstract Advanced stages of papillary and anaplastic thyroid cancer resistant cell lines. Interestingly, activation of the MAPK path- represent a highly aggressive subset, in which there are currently way was increased in all four of the dasatinib-resistant cell lines, few effective therapies. We and others have recently demon- likely due to B-Raf and c-Raf dimerization. Furthermore, strated that c-SRC is a key mediator of growth, invasion, and MAP2K1/MAP2K2 (MEK1/2) inhibition restored sensitivity in metastasis, and therefore represents a promising therapeutic all four of the dasatinib-resistant cell lines, and overcame target in thyroid cancer. However, clinically, Src inhibitor acquired resistance to dasatinib in the RAS-mutant Cal62 cell efficacy has been limited, and therefore further insights are line, in vivo. Together, these studies demonstrate that acquisi- needed to define resistance mechanisms and determine rational tion of the c-SRC gatekeeper mutation and MAPK pathway combination therapies. We have generated four thyroid cancer signaling play important roles in promoting resistance to the cell lines with a greater than 30-fold increase in acquired Src inhibitor dasatinib. We further demonstrate that up-front resistance to the Src inhibitor dasatinib.
    [Show full text]
  • Revisiting Clinical Trials Using EGFR Inhibitor-Based
    www.impactjournals.com/oncotarget/ Oncotarget, May, Vol.4, No 5 Revisiting Clinical Trials Using EGFR Inhibitor-Based Regimens in Patients with Advanced Non-Small Cell Lung Cancer: A Retrospective Analysis of an MD Anderson Cancer Center Phase I Population Jennifer Wheler1, Gerald Falchook1, Apostolia M. Tsimberidou1, David Hong1, Aung Naing1, Sarina Piha-Paul1, Su S. Chen2, John Heymach3, Siqing Fu1, Bettzy Stephen1, Jansina Y. Fok1, Filip Janku1, Razelle Kurzrock4 1 Department of Investigational Cancer Therapeutics – a Phase I Clinical Trials Program, The University of Texas MD Anderson Cancer Center, Texas 2 Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Texas 3 Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Texas 4 Moores Cancer Center, University of California, San Diego Correspondence to: Jennifer Wheler, email: [email protected] Keywords: EGFR mutation, EGFR wild-type, non-small cell lung cancer, phase I trials, resistance, squamous cell Received: May 10, 2013 Accepted: June 2, 2013 Published: June 4, 2013 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT: Purpose: Single-agent EGFR inhibitor therapy is effective mainly in patients with lung cancer and EGFR mutations. Treating patients who develop resistance, or who are insensitive from the outset, often because of resistant mutations, other aberrations or the lack of an EGFR mutation, probably requires rational combinations. We therefore investigated the outcome of EGFR inhibitor-based combination regimens in patients with heavily-pretreated non-small cell lung cancer (NSCLC) referred to a Phase I Clinic.
    [Show full text]
  • A Network Medicine Approach for Drug Repurposing in Duchenne Muscular Dystrophy
    G C A T T A C G G C A T genes Article A Network Medicine Approach for Drug Repurposing in Duchenne Muscular Dystrophy Salvo Danilo Lombardo 1 , Maria Sofia Basile 2 , Rosella Ciurleo 2, Alessia Bramanti 2, Antonio Arcidiacono 3, Katia Mangano 3 , Placido Bramanti 2, Ferdinando Nicoletti 3,* and Paolo Fagone 3 1 Department of Structural & Computational Biology at the Max Perutz Labs, University of Vienna, 1010 Vienna, Austria; [email protected] 2 IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; sofi[email protected] (M.S.B.); [email protected] (R.C.); [email protected] (A.B.); [email protected] (P.B.) 3 Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; [email protected] (A.A.); [email protected] (K.M.); [email protected] (P.F.) * Correspondence: [email protected] Abstract: Duchenne muscular dystrophy (DMD) is a progressive hereditary muscular disease caused by a lack of dystrophin, leading to membrane instability, cell damage, and inflammatory response. However, gene-editing alone is not enough to restore the healthy phenotype and additional treat- ments are required. In the present study, we have first conducted a meta-analysis of three microarray datasets, GSE38417, GSE3307, and GSE6011, to identify the differentially expressed genes (DEGs) be- tween healthy donors and DMD patients. We have then integrated this analysis with the knowledge Citation: Lombardo, S.D.; Basile, obtained from DisGeNET and DIAMOnD, a well-known algorithm for drug–gene association discov- M.S.; Ciurleo, R.; Bramanti, A.; eries in the human interactome.
    [Show full text]
  • Ado-Trastuzumab Emtansine: Drug Information
    Official reprint from UpToDate® www.uptodate.com ©2017 UpToDate® Ado-trastuzumab emtansine: Drug information Copyright 1978-2017 Lexicomp, Inc. All rights reserved. (For additional information see "Ado-trastuzumab emtansine: Patient drug information ") For abbreviations and symbols that may be used in Lexicomp (show table) ALERT: US Boxed Warning Do not interchange: Do not substitute ado-trastuzumab emtansine (Kadcyla) for or with trastuzumab (Herceptin). Hepatotoxicity: Serious hepatotoxicity has been reported, including liver failure and death. Monitor serum transaminases and bilirubin prior to initiation of treatment and prior to each dose. Reduce the dose or discontinue ado- trastuzumab emtansine as appropriate in cases of increased serum transaminases or total bilirubin. Cardiotoxicity: Ado-trastuzumab emtansine administration may lead to reductions in left ventricular ejection fraction (LVEF). Evaluate left ventricular function in all patients prior to and during treatment. Withhold treatment for a clinically significant decrease in left ventricular function. Pregnancy: Exposure to ado-trastuzumab emtansine during pregnancy can result in embryo-fetal harm. Advise patients of these risks and the need for effective contraception. Brand Names: US Kadcyla Brand Names: Canada Kadcyla Pharmacologic Category Antineoplastic Agent, Anti-HER2; Antineoplastic Agent, Antibody Drug Conjugate; Antineoplastic Agent, Antimicrotubular; Antineoplastic Agent, Monoclonal Antibody Dosing: Adult Note: Do not substitute ado-trastuzumab emtansine (US) or trastuzumab emtansine (Canada) for or with conventional trastuzumab; products are different and are NOT interchangeable. Breast cancer, metastatic, HER2+: IV: 3.6 mg/kg every 3 weeks until disease progression or unacceptable toxicity; Maximum dose: 3.6 mg/kg Missed or delayed doses: If a planned dose is missed or delayed, administer as soon as possible (at the dose and rate most recently tolerated), do not wait until the next planned cycle.
    [Show full text]
  • Targeted Therapy–Based Combination Treatment in Rhabdomyosarcoma Anke E.M
    Review Molecular Cancer Therapeutics Targeted Therapy–based Combination Treatment in Rhabdomyosarcoma Anke E.M. van Erp1, Yvonne M.H. Versleijen-Jonkers1, Winette T.A. van der Graaf1,2, and Emmy D.G. Fleuren3 Abstract Targeted therapies have revolutionized cancer treatment; in this regard, as this affects multiple hallmarks of cancer at however, progress lags behind in alveolar (ARMS) and embry- once. To determine the most promising and clinically relevant onal rhabdomyosarcoma (ERMS), a soft-tissue sarcoma mainly targeted therapy–based combination treatments for ARMS and occurring at pediatric and young adult age. Insulin-like growth ERMS, we provide an extensive overview of preclinical and factor 1 receptor (IGF1R)-directed targeted therapy is one of the (early) clinical data concerning a variety of targeted therapy– few single-agent treatments with clinical activity in these dis- based combination treatments. We concentrated on the most eases. However, clinical effects only occur in a small subset of common classes of targeted therapies investigated in rhabdo- patients and are often of short duration due to treatment myosarcoma to date, including those directed against receptor resistance. Rational selection of combination treatments of tyrosine kinases and associated downstream signaling path- either multiple targeted therapies or targeted therapies with ways, the Hedgehog signaling pathway, apoptosis pathway, chemotherapy could hypothetically circumvent treatment resis- DNA damage response, cell-cycle regulators, oncogenic fusion tance mechanisms and enhance clinical efficacy. Simultaneous proteins, and epigenetic modifiers. Mol Cancer Ther; 17(7); 1365–80. targeting of distinct mechanisms might be of particular interest Ó2018 AACR. Introduction treatment including surgery, chemotherapy, and radiotherapy has increased the 5-year overall survival (OS) to approximately Rhabdomyosarcoma is the most common type of soft-tissue 70%–90% for intermediate- and low-risk rhabdomyosarcoma, sarcoma (STS) observed in young patients with the most respectively.
    [Show full text]
  • Kinase Drug Discovery 20 Years After Imatinib: Progress and Future Directions
    REVIEWS Kinase drug discovery 20 years after imatinib: progress and future directions Philip Cohen 1 ✉ , Darren Cross 2 ✉ and Pasi A. Jänne 3 ✉ Abstract | Protein kinases regulate nearly all aspects of cell life, and alterations in their expression, or mutations in their genes, cause cancer and other diseases. Here, we review the remarkable progress made over the past 20 years in improving the potency and specificity of small-molecule inhibitors of protein and lipid kinases, resulting in the approval of more than 70 new drugs since imatinib was approved in 2001. These compounds have had a significant impact on the way in which we now treat cancers and non- cancerous conditions. We discuss how the challenge of drug resistance to kinase inhibitors is being met and the future of kinase drug discovery. Protein kinases In 2001, the first kinase inhibitor, imatinib, received FDA entered clinical trials in 1998, changed the perception Enzymes that catalyse transfer approval, providing the catalyst for an article with the of protein kinases as drug targets, which had previously of the γ- phosphate of ATP provocative title ‘Protein kinases — the major drug tar- received scepticism from many pharmaceutical com- to amino acid side chains in gets of the twenty- first century?’1. Imatinib inhibits the panies. Since then, hundreds of protein kinase inhibi- substrate proteins, such as serine, threonine and tyrosine Abelson (ABL) tyrosine kinase, which is expressed as a tors have been developed and tested in humans and, at residues. deregulated fusion protein, termed BCR–ABL, in nearly the time of writing, 76 have been approved for clinical all cases of chronic myeloid leukaemia (CML)2 and is use, mainly for the treatment of various cancers (FiG.
    [Show full text]