Targeting the FMS-Like Tyrosine Kinase 3 in Acute Myeloid Leukemia

Total Page:16

File Type:pdf, Size:1020Kb

Targeting the FMS-Like Tyrosine Kinase 3 in Acute Myeloid Leukemia Leukemia (2012) 26, 2176–2185 & 2012 Macmillan Publishers Limited All rights reserved 0887-6924/12 www.nature.com/leu REVIEW Targeting the FMS-like tyrosine kinase 3 in acute myeloid leukemia R Swords1, C Freeman2 and F Giles3 Acute myeloid leukemia (AML) is a highly heterogenous disease with multiple signaling pathways contributing to its pathogenesis. A key driver of AML is the FMS-like tyrosine kinase receptor-3 (FLT3). Activating mutations in FLT3, primarily the FLT3-internal tandem duplication (FLT3-ITD), are associated with decreased progression-free and overall survival. Identification of the importance of FLT3-ITD and the FLT3 pathway in the prognosis of patients with AML has stimulated efforts to develop therapeutic inhibitors of FLT3. Although these inhibitors have shown promising antileukemic activity, they have had limited efficacy to date as single agents and may require use in combination with cytotoxic chemotherapies. Here, we review clinical and preclinical results for the clinically mature FLT3 inhibitors currently in development. We conclude that multitargeted FLT3 inhibitors may have more utility earlier in the course of disease, when in vitro evidence suggests that AML cells are less dependent on FLT3 signaling, perhaps because of upregulation of multiple other signaling pathways. More potent agents may have greater utility in relapsed and heavily pretreated patients, in whom high levels of circulating FLT3 ligand may necessitate use of an agent with a very favorable pharmacokinetic/ pharmacodynamic profile. Novel combination regimens are also discussed. Leukemia (2012) 26, 2176–2185; doi:10.1038/leu.2012.114 Keywords: FLT3; tyrosine kinase inhibitors; acute myeloid leukemia; internal tandem duplication INTRODUCTION internal tandem duplication (ITD) in the FMS-like tyrosine kinase 10 Adult acute myeloid leukemia (AML) accounts for B30% of receptor-3 gene (FLT3), partial tandem duplication in the mixed 11 12 leukemias and 440% of leukemia-related deaths.1 AML is a highly lineage leukemia gene, Wilms tumor gene mutations and 13 heterogenous disease characterized by several karyotypic abnor- overexpression of the BAALC gene. Other mutations that may be malities (Table 1).2,3 Karyotypic abnormalities associated with a associated with poor prognosis in patients with AML include relatively good prognosis are present in B15% of patients 10–11-translocation oncogene family member 2, isocitrate and include translocation of chromosomes 15 and 17, t(15;17) dehydrogenase-1, isocitrate dehydrogenase-2 and additional 14 (q22;q21), translocation of chromosomes 8 and 21, t(8;21) sex comb-like 1. Alternatively, CCAAT/enhancer-binding (q22;q22) and inversion of chromosome 16, inv(16)(p13q22)/ protein a (CEBPA) and nucleophosmin 1 (NPM1) mutations (in t(16;16)(p13;q22). Prognostically bad karyotypic abnormalities, FLT3-ITD-negative patients) have been associated with improved 14,15 also present in B15% of patients, include deletion of chromo- prognosis. some 7, deletion of chromosome 5q and more than three In this review, we evaluate the prognostic implications of gene chromosomal abnormalities.4,5 Other karyotypic abnormalities mutations and cytogenetic risk factors in patients with AML, that have an unclear effect on prognosis have been observed, focusing on the FLT3-ITD mutation. We discuss established and including trisomy 8 and trisomy 21d. These abnormalities are emerging approaches using FLT3 inhibitors for the treatment generally considered to be in the intermediate-risk prognosis of AML. category along with normal karyotype. The identification of these markers led to the development of cytogenetic risk classification systems by several cooperative groups internationally, including the Cancer and Leukemia Group B,5 Eastern Cooperative Oncology STRUCTURE/FUNCTION OF THE FLT3 RECEPTOR and Southwest Oncology Groups,6 the UK Medical Research FLT3 receptor Council2 and Fro¨hling et al.4 in Germany. Mutations in FLT3 are among the most commonly occurring Most patients with AML have poor rates of survival, particularly and clinically relevant mutations in patients with AML. FLT3 is a those with a poor-risk karyotype. Five-year survival rates are receptor tyrosine kinase expressed on the leukemic blasts of B55%, 24% and 5% in patients with favorable-, intermediate- and 70–100% of patients with AML16,17 that is increasingly recognized poor-risk cytogenetics, respectively.5 Even among patients with as having a role in the pathogenesis of this disease. Upon binding normal karyotype AML, however, clinical outcomes are of the FLT3 ligand, FLT3 dimerizes and undergoes a conformational heterogenous. Among other patient-related factors, mutations in change whereby its activation loop opens, revealing the ATP- key genes involved in AML pathogenesis lead to large interpatient binding pocket. The receptor becomes autophosphorylated, variability in prognostic risk (Table 2).7–9 There is a plethora of inducing cell growth and inhibiting apoptosis through the mutations associated with poor prognosis in AML. These include activation of a signaling cascade involving proteins such as the 1Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA; 2Barts and The London NHS Trust, St Bartholomew’s Hospital, London, UK and 3HRB Clinical Research Facilities, National University of Ireland Galway and Trinity College Dublin, Dublin, Ireland. Correspondence: Dr FJ Giles, HRB Clinical Research Facility, National University of Ireland, University Road, Galway, Ireland. E-mail: [email protected] Received 28 December 2011; revised 22 March 2012; accepted 10 April 2012; accepted article preview online 27 April 2012; advance online publication, 22 May 2012 Role of FLT3 in AML R Swords et al 2177 Ras-GTPase-activating protein, phospholipase C, signal transducer Table 1. Karyotypic abnormalities in patients with AML, classified 18 5 and activator of transcription 5 and ERK1/2 (Figure 1). according to cytogenetic risk by the Cancer and Leukemia Group B, 19,20 Eastern Cooperative Oncology Group/Southwest Oncology Group6 Whereas, FLT3 ligand is ubiquitously expressed, the FLT3 and Medical Research Council3 receptor is localized primarily in hematopoietic and neural tissues.21 FLT3 is integral to hematopoiesis through its Cytogenetic risk group Abnormality Incidence, % regulation of proliferation, differentiation and apoptosis of hematopoietic progenitor cells. Knockout mice models have Favorable Inv(16) or t(16;16) 3.5–8.7 shown that lack of FLT3 ligand and FLT3 leads to reduced Del(9q) 1.6–2.8 hematopoietic precursors,22,23 demonstrating the importance of Del(16q) NR these proteins in hematopoietic expansion. t(8;21) 6.7–8.2 t(15;17) 4.4–12 FLT3 mutations Intermediate Normal karyotype 40–48 Several different mutations can occur in the FLT3 receptor in À 7 3.8–8.5 À Y 3.3–4.8 patients with AML. The most common of these is an ITD on exon Del(5q) 1.7–5.9 14 of the FLT3 gene, which varies from 3 to 4400 base pairs but is Del(7q) 1.6–8.5 typically in frame and occurs in the juxtamembrane region of the Del(9q) 1.6–2.8 receptor.24 The ITD disrupts the autoinhibitory function of the Del(12p) NR juxtamembrane domain, resulting in constitutive autophosphory- Del(20q) 1.3 lation of FLT3 and activation of its downstream effectors.25,26 þ 6NRThe FLT3-ITD mutation produces growth factor-independent þ 8 8.7–10.1 proliferation in leukemia cell lines and a fatal myeloproliferative 11 1.6 þ syndrome in murine models.27 Interestingly, however, when FLT3- þ 13 2.4 ITD was expressed in cells from knockout mice lacking FLT3 þ 21 2.3–2.8 28 t(6;9) 0.7–1.8 ligand, the receptor was only weakly activated. This suggests t(9;11) 2.2 that the ITD leads to gain of function mainly by inducing 11q23 3.7 hyperresponsivity of the FLT3 receptor to FLT3 ligand rather than through autoactivation of the receptor. a Unfavorable/adverse Complex 5.9–11.7 Activating mutations within the kinase domain of FLT3 are also Abnormal 3q 2.5 observed in patients with AML. The most common is mutation of Inv(3) or t(3;3) 1.0–2.0 the aspartate 835 (D835) mutation in the activation loop of the 5 1.6–5.9 À tyrosine kinase domain, which occurs in B7% of patients with À 7 3.8–8.5 29,30 Del(5q) 1.7–5.9 AML. Del(7q) 1.6–8.5 t(6;9) 0.7–1.8 t(9;22) 1.3 FLT3 IN THE CLINIC a It has become clear that the presence of FLT3-ITD, which is Abbreviations: AML, acute myeloid leukemia; NR, not reported. Complex 31,32 karyotype defined as X3 cytogenetic abnormalities. observed in B25% of patients with AML, is a key indicator of poor long-term prognosis. This has led to a concerted effort to Table 2. Prognostically relevant mutations in patients with AML Abnormality Prognostic outcome Study results P-value Negative patient outcome FLT3-ITD (compared with non-ITD FLT3) Shorter median OS15 10 vs 15.5 months 0.015 Shorter median DFS15 8 vs 12.5 months 0.033 MLL partial tandem duplication Shorter duration of CR117 7.1 vs 23.2 months 0.01 (compared with MLL WT) Shorter median OS11 5 vs 12 months 0.006 Shorter median RFS11 4 months vs NR o0.001 Shorter 2-year EFS118 20% vs 66% 0.03 Overexpression of BAALC (compared Shorter median OS13 1.7 vs 5.8 years 0.02 with low BAALC expression) Shorter median EFS13 0.8 vs 4.9 years 0.03 Shorter median DFS13 1.4 vs 7.3 years 0.03 Positive patient outcome Mutated CEBPA (compared with Longer median duration of CR46 NR vs 26 months 0.01 nonmutated CEBPA) Longer median OS46 NR vs 19 months 0.05 NPM1 mutations (FLT3-ITD À pts Higher rates of CR44 86% vs 63% o0.001a compared with FLT3-ITD þ pts) Longer median OS43 1183 vs 321 days 0.014 Longer median EFS43 773 vs 234 days 0.001 Abbreviations: AML, acute myeloid leukemia; CEBPA, CCAAT/enhancer-binding protein a; CR, complete response; DFS, disease-free survival; EFS, event-free survival; FLT3, FMS-like tyrosine kinase receptor-3 gene; ITD, internal tandem duplication; MLL, mixed lineage leukemia; NPM1, nucleophosmin 1; NR, not reached; OS, overall survival; Pts, patients; RFS, relapse-free survival; WT, wild-type.
Recommended publications
  • How I Treat Myelofibrosis
    From www.bloodjournal.org by guest on October 7, 2014. For personal use only. Prepublished online September 16, 2014; doi:10.1182/blood-2014-07-575373 How I treat myelofibrosis Francisco Cervantes Information about reproducing this article in parts or in its entirety may be found online at: http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests Information about ordering reprints may be found online at: http://www.bloodjournal.org/site/misc/rights.xhtml#reprints Information about subscriptions and ASH membership may be found online at: http://www.bloodjournal.org/site/subscriptions/index.xhtml Advance online articles have been peer reviewed and accepted for publication but have not yet appeared in the paper journal (edited, typeset versions may be posted when available prior to final publication). Advance online articles are citable and establish publication priority; they are indexed by PubMed from initial publication. Citations to Advance online articles must include digital object identifier (DOIs) and date of initial publication. Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036. Copyright 2011 by The American Society of Hematology; all rights reserved. From www.bloodjournal.org by guest on October 7, 2014. For personal use only. Blood First Edition Paper, prepublished online September 16, 2014; DOI 10.1182/blood-2014-07-575373 How I treat myelofibrosis By Francisco Cervantes, MD, PhD, Hematology Department, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain Correspondence: Francisco Cervantes, MD, Hematology Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain. Phone: +34 932275428.
    [Show full text]
  • Thrombopoietin Supports the Continuous Growth of Cytokine-Dependent Human Leukemia Cell Lines HG Drexler, M Zaborski and H Quentmeier
    Leukemia (1997) 11, 541–551 1997 Stockton Press All rights reserved 0887-6924/97 $12.00 Thrombopoietin supports the continuous growth of cytokine-dependent human leukemia cell lines HG Drexler, M Zaborski and H Quentmeier DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Cultures, Mascheroder Weg 1 B, D-38124 Braunschweig, Germany Hematopoiesis is a complex process of regulated cellular pro- nate membrane receptor. This binding triggers a series of intra- liferation and differentiation from the primitive stem cells to the cellular mediators involved in the growth factor’s signaling final fully differentiated cell. The long and extensive search for a factor specifically regulating megakaryocytopoiesis led to the pathways. Recently, a novel hematopoietic growth factor, cloning of a hormone, here called thrombopoietin (TPO), that termed thrombopoietin (TPO), was cloned and shown to be a specifically promotes proliferation and differentiation of the megakaryocytic lineage-associated growth and differentiation megakaryocytic lineage. The availability of recombinant TPO factor. Binding of TPO to its receptor, c-MPL, mediates plei- and its imminent clinical use has made a more detailed under- otropic effects on megakaryocyte development in vitro and in standing of its effects on hematopoietic cells more urgent. Nor- vivo. TPO is clearly the primary regulator of this cell lineage mal megakaryocyto- and thrombopoiesis occurs predomi- nantly in the bone marrow, a difficult organ to study in situ, acting at all levels of megakaryocytopoiesis and thrombopo- particularly in humans, due to the low numbers of megakary- iesis (reviewed in Ref. 1). ocytic progenitors and the consequent difficult isolation as The availability of TPO will be of considerable clinical pure populations.
    [Show full text]
  • Therapeutic Class Overview Colony Stimulating Factors
    Therapeutic Class Overview Colony Stimulating Factors Therapeutic Class Overview/Summary: This review will focus on the granulocyte colony stimulating factors (G-CSFs) and granulocyte- macrophage colony stimulating factors (GM-CSFs).1-5 Colony-stimulating factors (CSFs) fall under the naturally occurring glycoprotein cytokines, one of the main groups of immunomodulators.6 In general, these proteins are vital to the proliferation and differentiation of hematopoietic progenitor cells.6-8 The G- CSFs commercially available in the United States include pegfilgrastim (Neulasta®), filgrastim (Neupogen®), filgrastim-sndz (Zarxio®), and tbo-filgrastim (Granix®). While filgrastim-sndz and tbo- filgrastim are the same recombinant human G-CSF as filgrastim, only filgrastim-sndz is considered a biosimilar drug as it was approved through the biosimilar pathway. At the time tbo-filgrastim was approved, a regulatory pathway for biosimilar drugs had not yet been established in the United States and tbo-filgrastim was filed under its own Biologic License Application.9 Only one GM-CSF is currently available, sargramostim (Leukine). These agents are Food and Drug Administration (FDA)-approved for a variety of conditions relating to neutropenia or for the collection of hematopoietic progenitor cells for collection by leukapheresis.1-5 Due to the pathway taken, tbo-filgrastim does not share all of the same indications as filgrastim and these two products are not interchangeable. It is important to note that although filgrastim-sndz is a biosimilar product, and it was approved with all the same indications as filgrastim at the time, filgrastim has since received FDA-approval for an additional indication that filgrastim-sndz does not have, to increase survival in patients with acute exposure to myelosuppressive doses of radiation.1-3A complete list of indications for each agent can be found in Table 1.
    [Show full text]
  • Side Effects of Molecular-Targeted Therapies in Solid Cancers : a New Challenge in Cancer Therapy Management
    Side effects of molecular-targeted therapies in solid cancers : a new challenge in cancer therapy management Ahmad Awada, MD, PhD Medical Oncology Clinic Institut Jules Bordet Université Libre de Bruxelles (U.L.B.) Brussels, Belgium PLAN OF THE LECTURE 1. Concept 2. Achievements on the management of side effects 3. Remaining challenges 4. New challenges with the development of molecular-targeted therapies 5. Conclusions Reducing the cancer- related problems and the side effects of the SUPPORTIVE CARE = medicine administered to treat the disease SIDE EFFECTS OF CANCER THERAPY: ACHIEVEMENTS Side effect Preventive & Therapeutic intervention • Febrile neutropenia • G-CSF, Anti-infectives • Anemia • Epoetine •Mucositis •Laser therapy, Palifermin • Nausea & Vomiting • 5-HT3 and neurokin-1-receptor antagonists •Thromboembolic •LMW Heparin events • Cardiomyopathy • Liposomal formulations, Dexrazonane (anthracyclines) MANAGEMENT OF SIDE EFFECTS : REMAINING CHALLENGES • Alopecia • Thrombocytopenia ( ! Promising Thrombopoietin- mimetics are under investigation) • Asthenia MOLECULAR TARGETS AND THERAPIES (1) Drug Class Mechanism of action Main tumor indication Gefitinib* Small molecule TK inhibitor of EGFR NSCLC (Iressa) Erlotinib* Small molecule TK inhibitor of EGFR NSCLC (Tarceva) Cetuximab* Monoclonal Antibody Blocks EGFR Colorectal, Head & (Erbitux) Neck, NSCLC Monoclonal Panitumumab* Antibody Blocks EGFR Colorectal (Vectibix) * Investigational in BC TK : tyrosine kinase; EGFR : epidermal growth factor receptor MOLECULAR TARGETS AND THERAPIES
    [Show full text]
  • DOPTELET (Avatrombopag) RATIONALE for INCLUSION IN
    DOPTELET (avatrombopag) RATIONALE FOR INCLUSION IN PA PROGRAM Background Doptelet is a thrombopoietin (TPO) receptor agonist used to increase platelet counts. Doptelet (avatrombopag) is an orally bioavailable, small molecule TPO receptor agonist that stimulates proliferation and differentiation of megakaryocytes from bone marrow progenitor cells resulting in an increased production of platelets. Doptelet does not compete with TPO for binding to the TPO receptor and has an additive effect with TPO on platelet production (1). Regulatory Status FDA approved indication: Doptelet is a thrombopoietin receptor agonist indicated for the treatment of: (1) 1. Thrombocytopenia in adult patients with chronic liver disease who are scheduled to undergo a procedure. 2. Thrombocytopenia in adult patients with chronic immune thrombocytopenia who have had an insufficient response to a previous treatment. Doptelet should not be administered to patients with chronic liver disease in an attempt to normalize platelet counts (1). Doptelet is a thrombopoietin (TPO) receptor agonist and TPO receptor agonists have been associated with thrombotic and thromboembolic complications in patients with chronic liver disease. A Doppler ultrasound is a noninvasive test that can be used to estimate the blood flow through blood vessels by bouncing high-frequency sound waves (ultrasound) off circulating red blood cells. A Doppler ultrasound may help determine if Doptelet therapy is appropriate for a patient (1-2). The safety and effectiveness of Doptelet in pediatric patients have not been established (1). Summary Doptelet is a thrombopoietin (TPO) receptor agonist used to increase platelet counts. Doptelet (avatrombopag) is an orally bioavailable, small molecule TPO receptor agonist that stimulates proliferation and differentiation of megakaryocytes from bone marrow progenitor cells resulting in an increased production of platelets.
    [Show full text]
  • Insights Into the Cellular Mechanisms of Erythropoietin-Thrombopoietin Synergy
    Papayannopoulou et al.: Epo and Tpo Synergy Experimental Hematology 24:660-669 (19961 661 @ 1996 International Society for Experimental Hematology Rapid Communication ulation with fluorescence microscopy. Purified subsets were grown in plasma clot and methylcellulose clonal cultures and in suspension cultures using the combinations of cytokines Insights into the cellular mechanisms cadaveric bone marrow cells obtained from Northwest described in the text. Single cells from the different subsets Center, Puget Sound Blood Bank (Seattle, WA), were were. also deposited (by FACS) on 96-well plates containing of erythropoietin-thrombopoietin synergy washed, and incubated overnight in IMDM with 10% medmm and cytokines. Clonal growth from single-cell wells calf serum on tissue culture plates to remove adherent were double-labeled with antiglycophorin A-PE and anti­ Thalia Papayannopoulou, Martha Brice, Denise Farrer, Kenneth Kaushansky From the nonadherent cells, CD34+ cells were isolated CD41- FITC between days 10 and 19. direct immunoadherence on anti-CD34 monoclonal anti­ University of Washington, Department of Medicine, Seattle, WA (mAb)-coated plates, as previously described [15]. Purity Immunocytochemistry Offprint requests to: Thalia Papayannopoulou, MD, DrSci, University of Washington, isolated CD34+ cells ranged from 80 to 96% by this For immunocytochemistry, either plasma clot or cytospin cell Division of Hematology, Box 357710, Seattle, WA 98195-7710 od. Peripheral blood CD34 + cells from granulocyte preparations were used. These were fixed at days 6-7 and (Received 24 January 1996; revised 14 February 1996; accepted 16 February 1996) ulating factor (G-CSF)-mobilized normal donors 12-13 with pH 6.5 Histochoice (Amresco, Solon, OH) and provided by Dr.
    [Show full text]
  • FLT3 Inhibitors in Acute Myeloid Leukemia Mei Wu1, Chuntuan Li2 and Xiongpeng Zhu2*
    Wu et al. Journal of Hematology & Oncology (2018) 11:133 https://doi.org/10.1186/s13045-018-0675-4 REVIEW Open Access FLT3 inhibitors in acute myeloid leukemia Mei Wu1, Chuntuan Li2 and Xiongpeng Zhu2* Abstract FLT3 mutations are one of the most common findings in acute myeloid leukemia (AML). FLT3 inhibitors have been in active clinical development. Midostaurin as the first-in-class FLT3 inhibitor has been approved for treatment of patients with FLT3-mutated AML. In this review, we summarized the preclinical and clinical studies on new FLT3 inhibitors, including sorafenib, lestaurtinib, sunitinib, tandutinib, quizartinib, midostaurin, gilteritinib, crenolanib, cabozantinib, Sel24-B489, G-749, AMG 925, TTT-3002, and FF-10101. New generation FLT3 inhibitors and combination therapies may overcome resistance to first-generation agents. Keywords: FMS-like tyrosine kinase 3 inhibitors, Acute myeloid leukemia, Midostaurin, FLT3 Introduction RAS, MEK, and PI3K/AKT pathways [10], and ultim- Acute myeloid leukemia (AML) remains a highly resist- ately causes suppression of apoptosis and differentiation ant disease to conventional chemotherapy, with a me- of leukemic cells, including dysregulation of leukemic dian survival of only 4 months for relapsed and/or cell proliferation [11]. refractory disease [1]. Molecular profiling by PCR and Multiple FLT3 inhibitors are in clinical trials for treat- next-generation sequencing has revealed a variety of re- ing patients with FLT3/ITD-mutated AML. In this re- current gene mutations [2–4]. New agents are rapidly view, we summarized the preclinical and clinical studies emerging as targeted therapy for high-risk AML [5, 6]. on new FLT3 inhibitors, including sorafenib, lestaurtinib, In 1996, FMS-like tyrosine kinase 3/internal tandem du- sunitinib, tandutinib, quizartinib, midostaurin, gilteriti- plication (FLT3/ITD) was first recognized as a frequently nib, crenolanib, cabozantinib, Sel24-B489, G-749, AMG mutated gene in AML [7].
    [Show full text]
  • Comparison Between Filgrastim and Lenograstim Plus Chemotherapy For
    Bone Marrow Transplantation (2010) 45, 277–281 & 2010 Macmillan Publishers Limited All rights reserved 0268-3369/10 $32.00 www.nature.com/bmt ORIGINAL ARTICLE Comparison between filgrastim and lenograstim plus chemotherapy for mobilization of PBPCs R Ria, T Gasparre, G Mangialardi, A Bruno, G Iodice, A Vacca and F Dammacco Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy Recombinant human (rHu) G-CSF has been widely used during transplantation.4 However, the use of G-CSF after to treat neutropenia and mobilize PBPCs for their autologous PBPC transplantation has been queried, as its autologous and allogeneic transplantation. It shortens further reduction in time to a safe neutrophil count5,6 does neutropenia and thus reduces the frequency of neutropenic not always imply fewer significant clinical events, such as fever. We compared the efficiency of glycosylated rHu infections, length of hospitalization, extrahematological and non-glycosylated Hu G-CSF in mobilizing hemato- toxicities or mortality.7,8 Even so, the ASCO guidelines still poietic progenitor cells (HPCs). In total, 86 patients were recommend the use of growth factors after autologous consecutively enrolled for mobilization with CY plus either PBPC transplantation.9 glycosylated or non-glycosylated G-CSF, and under- G-CSF induces the proliferation and differentiation of went leukapheresis. The HPC content of each collection, myeloid precursor cells, and also provides a functional toxicity, days of leukapheresis needed to reach the activity that influences chemotaxis, respiratory burst and minimum HPC target and days to recover WBC (X500 Ag expression of neutrophils.
    [Show full text]
  • Transient Exposure to Quizartinib Mediates Sustained Inhibition of FLT3 Signaling While Specifically Inducing Apoptosis in FLT3-Activated Leukemia Cells
    Published OnlineFirst February 14, 2013; DOI: 10.1158/1535-7163.MCT-12-0305 Molecular Cancer Cancer Therapeutics Insights Therapeutics Transient Exposure to Quizartinib Mediates Sustained Inhibition of FLT3 Signaling while Specifically Inducing Apoptosis in FLT3-Activated Leukemia Cells Ruwanthi N. Gunawardane, Ronald R. Nepomuceno, Allison M. Rooks, Jeremy P. Hunt, Jill M. Ricono, Barbara Belli, and Robert C. Armstrong Abstract Fms-like tyrosine kinase 3 (FLT3) is implicated in the pathogenesis of acute myeloid leukemia (AML). FLT3-activating internal tandem duplication (ITD) mutations are found in approximately 30% of patients with AML and are associated with poor outcome in this patient population. Quizartinib (AC220) has previously been shown to be a potent and selective FLT3 inhibitor. In the current study, we expand on previous observations by showing that quizartinib potently inhibits the phosphorylation of FLT3 and downstream signaling molecules independent of FLT3 genotype, yet induces loss of viability only in cells expressing constitutively activated FLT3. We further show that transient exposure to quizartinib, whether in vitro or in vivo, leads to prolonged inhibition of FLT3 signaling, induction of apoptosis, and drastic reductions in tumor volume and pharmacodynamic endpoints. In vitro experiments suggest that these prolonged effects are mediated by slow binding kinetics that provide for durable inhibition of the kinase following drug removal/clearance. Together these data suggest quizartinib, with its unique combination of selectivity and potent/sustained inhibition of FLT3, may provide a safe and effective treatment against FLT3-driven leukemia. Mol Cancer Ther; 12(4); 1–10. Ó2013 AACR. Introduction downstream signaling cascades including the Ras/ Fms-like tyrosine kinase 3 (FLT3) is a member of the MAPK, PI3K/Akt, and STAT5 pathways (1, 9–13).
    [Show full text]
  • Letters to the Editor
    LETTERS TO THE EDITOR FLT3 inhibitor, sorafenib, induced no myelosuppression.5,6 Inhibition of c-Kit by tyrosine kinase inhibitors To better understand the relationship between inhibition of c-Kit, FLT3, and marrow suppression, we studied a series of different TKIs using bone marrow progenitor cell assays Several small molecule tyrosine kinase inhibitors (TKIs) and immunoblots. inhibit c-Kit, an effect associated with myelosuppression Cell lines were cultured as previously described.2 TF-1 and hair depigmentation. We studied a panel of approved cells were obtained from the American Type Culture and investigational TKIs for inhibitory activity against FLT3 Collection (ATCC; Manassas, VA, USA) and grown in and c-Kit, and on hematopoietic progenitor cells. Potent c- RPMI supplemented with GM-CSF (Invitrogen, Grand Kit inhibitors such as dasatinib, pazopanib, and quizartinib Island, NY, USA). Quizartinib was obtained from Ambit demonstrated the greatest disruption of hematopoietic pro- Biosciences (San Diego, CA, USA). Crenolanib was genitor cells, while sorafenib, which has negligible activity obtained from Arog Pharmaceuticals (Dallas, TX, USA). against c-Kit, demonstrated only minimal disruption. Our Dasatinib, pazopanib, and imatinib were obtained from LC data highlight the importance of determining a therapeutic Laboratories (Woburn, MA, USA). Electrophoresis, index between the targeted receptor and c-Kit for TKIs immunoblotting, and hematopoietic progenitor cell assays used to treat malignancies in order to maintain normal were performed as described.2 Cytokines used included hematopoiesis and improve outcomes. SCF, G-CSF, GM-CSF, IL-3, IL-6, and erythropoietin. Myelosuppression is a common adverse event in new Unused portions of bone marrow from normal donors drug development in oncology.
    [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]
  • Human G-CSF ELISA Kit (ARG80143)
    Product datasheet [email protected] ARG80143 Package: 96 wells Human G-CSF ELISA Kit Store at: 4°C Component Cat. No. Component Name Package Temp ARG80143-001 Antibody-coated 8 X 12 strips 4°C. Unused strips microplate should be sealed tightly in the air-tight pouch. ARG80143-002 Standard 3 X 2 ng/vial 4°C (Lyophilized) ARG80143-003 Standard diluent 20 ml 4°C buffer ARG80143-004 Antibody conjugate 400 µl 4°C concentrate ARG80143-005 Antibody diluent 16 ml 4°C buffer ARG80143-006 HRP-Streptavidin 400 µl 4°C (Protect from concentrate light) ARG80143-007 HRP-Streptavidin 16 ml 4°C diluent buffer ARG80143-008 20X Wash buffer 50 ml 4°C ARG80143-009 TMB substrate 12ml 4°C (Protect from light) ARG80143-010 STOP solution 12ml 4°C ARG80143-011 Plate sealer 4 strips Room temperature Summary Product Description ARG80143 Human G-CSF ELISA Kit is an Enzyme Immunoassay kit for the quantification of Human G-CSF in Serum, Plasma, Cell culture supernatants. Tested Reactivity Hu Tested Application ELISA Specificity No significant cross-reactivity or interference with Human CT-1,IL-11,OSM,CNTF,HGF,M-CSF;mouse IL-6;rat IL-6,CNTF Target Name G-CSF Conjugation HRP Conjugation Note Substrate: TMB and read at 450 nm Sensitivity 15 pg/ml Sample Type Serum, Plasma, Cell culture supernatants Standard Range 31.25 - 2000 pg/ml www.arigobio.com 1/3 Sample Volume 100 µl Precision CV: Alternate Names Granulocyte colony-stimulating factor; Lenograstim; C17orf33; GCSF; G-CSF; Filgrastim; Pluripoietin; CSF3OS Application Instructions Assay Time 4 hours Properties Form 96 well Storage instruction Store the kit at 2-8°C.
    [Show full text]