FLT3 Inhibition: a Moving and Evolving Target in Acute Myeloid Leukaemia

Total Page:16

File Type:pdf, Size:1020Kb

FLT3 Inhibition: a Moving and Evolving Target in Acute Myeloid Leukaemia Leukemia (2013) 27, 260–268 & 2013 Macmillan Publishers Limited All rights reserved 0887-6924/13 www.nature.com/leu REVIEW FLT3 inhibition: a moving and evolving target in acute myeloid leukaemia AYH Leung, C-H Man and Y-L Kwong Internal tandem duplication (ITD) of the fms-like tyrosine kinase 3 (FLT3) gene is a gain-of-function mutation common in acute myeloid leukaemia (AML). It is associated with inferior prognosis and response to chemotherapy. Single base mutations at the FLT3 tyrosine kinase domain (TKD) also leads to a gain of function, although its prognostic significance is less well defined because of its rarity. The clinical benefits of FLT3 inhibition are generally limited to AML with FLT3-ITD. However, responses are transient and leukaemia progression invariably occurs. There is compelling evidence that leukaemia clones carrying both ITD and TKD mutations appear when resistance to FLT3 inhibitors occurs. Interestingly, the emergence of double ITD and TKD mutants can be recapitulated in vitro when FLT3-ITD þ leukaemia cell lines are treated with mutagens and FLT3 inhibitors. Furthermore, murine xenotransplantation models also suggest that, in some cases, the FTL3-ITD and TKD double mutants actually exist in minute amounts before treatment with FLT3 inhibitors, expand under the selection pressure of FLT3 inhibition and become the predominant resistant clone(s) during the drug-refractory phase. On the basis of this model of clonal evolution, a multipronged strategy using more potent FLT3 inhibitors, and a combinatorial approach targeting both FLT3-dependent and FLT3-independent pathways, will be needed to improve outcome. Leukemia (2013) 27, 260–268; doi:10.1038/leu.2012.195 Keywords: FLT3; internal tandem duplication; tyrosine kinase domain mutation; inhibition INTRODUCTION Clinically, FLT3-ITD occurs most frequently in AML with normal 2,3 Acute myeloid leukaemia (AML) is a group of heterogeneous karyotype, trisomy 8, t(6;9) and t(15;17), and is generally 17 diseases sharing in common an abnormal increase in myeloblasts associated with leukocytosis on presentation. Although the in the circulation and bone marrow. Intensive chemotherapy and complete remission (CR) rate does not appear to be affected, 18 allogeneic haematopoietic stem cell (HSC) transplantation are the AMLs with FLT3-ITD have higher relapse rates and therefore 19 principal treatment modalities. However, these approaches have inferior disease-free and overall survivals, particularly in AML 20 21 22 reached an impasse, with a cure rate of only 30–40%.1 Internal with larger ITD size, higher allelic burden and multiple ITDs. tandem duplication (ITD) of the fms-like tyrosine kinase 3 (FLT3) Therefore, FLT3 inhibition has become a legitimate therapeutic gene is a gain-of-function mutation, and represents one of the option, and clinical trials of FLT3 inhibitors in AML have been 23–25 most common genetic alterations in AML, occurring in nearly 30% ongoing for a decade. To date, more than 20 small molecule of cases.2,3 FLT3 is a class III receptor tyrosine kinase consisting of inhibitors against FLT3 have been reported, of which eight of 26 five extracellular immunoglobulin-like domains, a transmembrane them have been evaluated in clinical trials. Structurally, most of domain, a juxtamembrane domain and two tyrosine kinase these inhibitors are heterocyclic compounds, inhibiting FLT3 domains (TKD1 and TKD2).4 It is highly expressed in haemato- activity by competing with ATP for binding to the ATP-binding poietic stem and progenitor cells.5 Upon binding to FLT3 ligand, it pocket of the TKD. Functionally, they are generally multikinase dimerizes and autophosphorylates, resulting in activation of inhibitors. Their clinical activities appear to be mediated by FLT3 tyrosine kinase activity, which in turn activates the phosphoino- inhibition, so that their efficacies are limited to AML carrying FLT3- sitide-3-kinase/AKT and RAS/mitogen-activated protein kinase ITD, and correlated with inhibition of FLT3 phosphorylation and 27–31 pathways.6,7 FLT3-ITD involves an in-frame duplication of 3–400 hence its downstream signalling effectors. Although basepairs at the juxtamembrane or TKD1 domains. It results inhibition of FLT3 may be achievable, clinical efficacy is less than in alteration of cellular signalling, including the constitutive convincing, being limited by the invariable leukaemia progression 24 activation of FLT3 independent of ligand binding;8 activation of despite continuous treatment. Comprehensive reviews on 24,32–39 STAT5 (signal transducer and activator of transcription 5) via SRC the mechanisms of drug resistance have been published. kinase;9 phosphorylation of a transcription factor FOXO3A;10 The proposed mechanisms include persistent activation of FLT3 downregulation of the equilibrative nucleoside transporter 1 for signalling due to overexpression of FLT3 ligand and protein, cytarabine;11 and induction of reactive oxygen species production.12 activation of antiapoptotic signals and protection of leukaemia- These molecular consequences result in damage13 and defective initiating cells (LIC) by the bone marrow niche (Table 1). Although repair of DNA, and increased cellular proliferation and resistance to therapeutic strategies have been formulated on these proposi- apoptosis.14–16 Comprehensive description of normal and aberrant tions, they are based primarily on in vitro data and laboratory FLT3 signalling has been reviewed previously.4,14–16 observations. Division of Haematology and Bone Marrow Transplantation, Department of Medicine, Queen Mary Hospital, Hong Kong, China. Correspondence: Professor Y-L Kwong, Division of Haematology and Bone Marrow Transplantation, Department of Medicine, Professorial Block, Queen Mary Hospital, Pokfulam Road, Hong Kong, China. E-mail: [email protected] Received 11 May 2012; revised 26 June 2012; accepted 6 July 2012; accepted article preview online 16 July 2012; advance online publication, 3 August 2012 FLT3 inhibition AYH Leung et al 261 Table 1. Proposed mechanisms of resistance to FLT3 inhibitors Proposed mechanisms Proposed means of targeting References Overexpression of FLT3 ligands Targeting FLT3 ligand as a therapeutic strategy Sato et al.34 Overexpression of FLT3 protein Degrading FLT3 proteins by inhibition of chaperone heat-shock protein 90 Weisberg et al.37, Nishioka et al.81 and Al Shaer et al.82 Protection by bone marrow niche Reducing CXCL12 signalling in stromal cells by the HDM2 antagonist Nutlin-3a Kojima et al.32 via stabilization of p53 Disrupting interaction between LSCs and niche (for example, CXCR4 antagonists) Parmar et al.33 Over-riding microenvironment-mediated resistance to FLT3 inhibitors by combination Weisberg et al.83 treatment with dasatinib and JAK inhibitors Overexpression of antiapoptotic genes Targeting STAT pathway or survivin with inhibitors or shRNA Zhou et al.38 Targeting MCL-1 to restore sensitivity to FLT3 inhibitors Breitenbuecher et al.39 and Sto¨lzel et al.35 Emergence of resistant clones See text Abbreviations: FLT3, fms-like tyrosine kinase 3; JAK, janus kinase; LSC, leukaemia stem cells; shRNA, short hairpin RNA; STAT, signal transducer and activator of transcription. Table 2. Clinical trials of FLT3 inhibitors in FLT3-mutated AML patients Inhibitor Dosing schedulea Other drugs Number of patients Outcome References ITD TKD mutation Lestaurtinib 40–60 mg b.i.d. None 16 1 BR: 30%; HI: 16% Smith et al.30 Lestaurtinib 60 mg b.i.d., days 1–28 None 2 3 BR: 60%; HI: 60% Knapper et al.84 Lestaurtinib 80 mg b.i.d., days 7–112 Mitoxantrone, etoposide 103b 11b No improvement in survival Levis et al.49 cytarabine, days 1–5 Midostaurin 75 mg t.i.d. None 18 2 CR: 5%; BR: 35%; HI: 30% Stone et al.85 Midostaurin 50 mg b.i.d. None 12 6 BR: 67%; HI: 50% Fischer et al.28 Sunitinib 50–75 mg daily, days 1–28 None 2 2 CRi: 25%; PR: 75% Fiedler et al.51 80 mg b.i.d., days 29–58 Tandutinib 150–700 mg b.i.d. None 8 1 BR: 25%; HI: 25% DeAngelo et al.50 KW-2449 25–500 mg b.i.d., days 1–14 None 11 0 BR: 45% Pratz et al.52 Sorafenib 600 mg daily–400 mg b.i.d. None 7c 3b CRi: 11%; BR: 56% Zhang et al.53 Sorafenib 200–800 mg daily None 6 0 CR: 33%; BR: 66% Metzelder et al.86 Sorafenib 400–600 mg b.i.d., 14–28 days None 2 0 BR: 50% Pratz et al.87 Sorafenib 400 mg b.i.d., induction: 7 days; Idarubicin, cytarabine 13 2 CR þ CRp: 100% Ravandi et al.55 maintenance: 28 days AC220 200 mg daily None 62 0 CR þ CRp þ CRi: 45% Cortes et al.54 Sorafenib 400 mg b.i.d. None 13 0 CRi: 46%, nCRi: 46% Man et al.61 Sorafenib 400 mg b.i.d. None 65 0 CR þ CRi þ CMR: 38% Metzelder et al.88 HI þ BR þ PR: 61% Midostaurin 50 mg b.i.d. Daunorubicin Cytarabine 9 4 CR: 92% Stone et al. 56 Abbreviations: AML, acute myeloid leukaemia; b.i.d., twice daily; t.i.d., three times daily; BR, blast response; CMR, complete molecular response; CR, complete remission; CRp, CR without platelet recovery; CRi, CR with incomplete haematological recovery; FLT3, fms-like tyrosine kinase 3; HI, haematological improvement; ITD, internal tandem duplication; nCRi, near CRi but with focal blast prominence in trephine biopsy that could not be enumerated; PR, partial remission; TKD, tyrosine kinase domain. aUnless stated, continued until maximal response or unacceptable toxicity. All medications were orally administered. bTwo patients had both ITD and TKD mutation. cOne patient had both ITD and TKD mutation. There is now compelling clinical evidence that the resistance to full-blown AML when additional genetic alterations, such as FLT3 inhibitors may arise from the emergence of FLT3-ITD þ NUP98-HoxD13, are present.47 These data can be considered as clones carrying additional TKD mutations.
Recommended publications
  • Stanford Chem-H Presentation (PDF)
    KiNativ® In situ kinase profiling Stanford University ChEM-H confidential @KiNativPlatform Principle of the KiNativ platform • ATP (or ADP) acyl phosphate binds to, and covalently modifies Lysine residues in the active site • Thus, ATP acyl phosphate with a desthiobiotin tag can be used capture and quantitate kinases in a complex lysate Acyl phosphate Desthiobiotin tag ATP 2 ATP acyl phosphate probe covalently modifies kinase in the active site Lysine 2 Lysine 1 3 ATP acyl phosphate probe covalently modifies kinase in the active site Lysine 2 Lysine 1 4 Samples trypsinized, probe-labeled peptides captured with streptavidin, and analyzed by targeted LC-MS2 Identification Quantitation Explicit determination of peptide Integration of signal from MS2 sequence and probe modification site fragment ions from MS2 spectrum 5 Comprehensive Coverage of Protein and Lipid Kinases Protein kinases Atypical kinases Green: Kinases detected on KiNativ Red: Kinases not detected on KiNativ ~80% of known protein and atypical kinases identified on the platform http://www.kinativ.com/coverage/protein-lipid.html 6 Profiling compound(s) on the KiNativ platform Control sample – add probe Sample: Lysate derived from any cell line or tissue from ANY species Treated sample – add inhibitor followed by probe Inhibited kinase Green: Kinases Blue: Probe Gray: Non-kinases Red: Inhibitor 7 Profiling compound(s) on the KiNativ platform Control sample – add probe MS signalMS Sample: Lysate derived from any cell line or tissue from ANY species Treated sample – add inhibitor
    [Show full text]
  • Download Product Insert (PDF)
    PRODUCT INFORMATION Pacritinib Item No. 16709 CAS Registry No.: 937272-79-2 Formal Name: 11-[2-(1-pyrrolidinyl)ethoxy]-14,19-dioxa- 5,7,27-triazatetracyclo[19.3.1.12,6.18,12] O heptacosa-1(25),2,4,6(27),8,10,12(26), 16E,21,23-decaene Synonym: SB1518 N N H MF: C28H32N4O3 FW: 472.6 N Purity: ≥98% O Stability: ≥2 years at -20°C Supplied as: A crystalline solid N O UV/Vis.: λmax: 285 nm Laboratory Procedures For long term storage, we suggest that pacritinib​ be stored as supplied at -20°C. It should be stable for at least two years. Pacritinib is supplied as a crystalline solid. A stock solution may be made by dissolving the pacritinib in the solvent of choice. Pacritinib is soluble in the organic solvent DMSO, which should be purged with an inert gas, at a concentration of approximately 0.5 mg/ml (slightly warmed). Pacritinib is sparingly soluble in aqueous solutions. To enhance aqueous solubility, dilute the organic solvent solution into aqueous buffers or isotonic saline. If performing biological experiments, ensure the residual amount of organic solvent is insignificant, since organic solvents may have physiological effects at low concentrations. We do not recommend storing the aqueous solution for more than one day. Description FMS-like tyrosine kinase 3 (FLT3) and Janus kinase 2 (JAK2) are tyrosine kinases that mediate cytokine signaling and are frequently mutated in cancers, particularly acute myeloid leukemia.1,2 Pacritinib is an 1 inhibitor of both FLT3 and JAK2 (IC50s = 22 and 23 nM, respectively).
    [Show full text]
  • 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]
  • Federal Register Notice 5-1-2020 Pdf Icon[PDF – 358
    Federal Register / Vol. 85, No. 85 / Friday, May 1, 2020 / Notices 25439 confidential by the respondent (5 U.S.C. schedules. Other than examination DEPARTMENT OF HEALTH AND 552(b)(4)). reports, it provides the only financial HUMAN SERVICES Current actions: The Board has data available for these corporations. temporarily revised the instructions to The Federal Reserve is solely Centers for Disease Control and the FR Y–9C report to accurately reflect responsible for authorizing, supervising, Prevention the revised definition of ‘‘savings and assigning ratings to Edges. The [CDC–2020–0046; NIOSH–233–C] deposits’’ in accordance with the Federal Reserve uses the data collected amendments to Regulation D in the on the FR 2886b to identify present and Hazardous Drugs: Draft NIOSH List of interim final rule published on April 28, potential problems and monitor and Hazardous Drugs in Healthcare 2020 (85 FR 23445). Specifically, the develop a better understanding of Settings, 2020; Procedures; and Risk Board has temporarily revised the activities within the industry. Management Information instructions on the FR Y–9C, Schedule HC–E, items 1(b), 1(c), 2(c) and glossary Legal authorization and AGENCY: Centers for Disease Control and content to remove the transfer or confidentiality: Sections 25 and 25A of Prevention, HHS. withdrawal limit. As a result of the the Federal Reserve Act authorize the ACTION: Notice and request for comment. revision, if a depository institution Federal Reserve to collect the FR 2886b chooses to suspend enforcement of the (12 U.S.C. 602, 625). The obligation to SUMMARY: The National Institute for six transfer limit on a ‘‘savings deposit,’’ report this information is mandatory.
    [Show full text]
  • Tanibirumab (CUI C3490677) Add to Cart
    5/17/2018 NCI Metathesaurus Contains Exact Match Begins With Name Code Property Relationship Source ALL Advanced Search NCIm Version: 201706 Version 2.8 (using LexEVS 6.5) Home | NCIt Hierarchy | Sources | Help Suggest changes to this concept Tanibirumab (CUI C3490677) Add to Cart Table of Contents Terms & Properties Synonym Details Relationships By Source Terms & Properties Concept Unique Identifier (CUI): C3490677 NCI Thesaurus Code: C102877 (see NCI Thesaurus info) Semantic Type: Immunologic Factor Semantic Type: Amino Acid, Peptide, or Protein Semantic Type: Pharmacologic Substance NCIt Definition: A fully human monoclonal antibody targeting the vascular endothelial growth factor receptor 2 (VEGFR2), with potential antiangiogenic activity. Upon administration, tanibirumab specifically binds to VEGFR2, thereby preventing the binding of its ligand VEGF. This may result in the inhibition of tumor angiogenesis and a decrease in tumor nutrient supply. VEGFR2 is a pro-angiogenic growth factor receptor tyrosine kinase expressed by endothelial cells, while VEGF is overexpressed in many tumors and is correlated to tumor progression. PDQ Definition: A fully human monoclonal antibody targeting the vascular endothelial growth factor receptor 2 (VEGFR2), with potential antiangiogenic activity. Upon administration, tanibirumab specifically binds to VEGFR2, thereby preventing the binding of its ligand VEGF. This may result in the inhibition of tumor angiogenesis and a decrease in tumor nutrient supply. VEGFR2 is a pro-angiogenic growth factor receptor
    [Show full text]
  • Efficacy and Safety of Midostaurin-Based Induction and Maintenance Therapy for Newly Diagnosed AML
    POST-ASH Issue 4, 2016 Efficacy and Safety of Midostaurin-Based Induction and Maintenance Therapy for Newly Diagnosed AML For more visit ResearchToPractice.com/5MJCASH2016 CME INFORMATION OVERVIEW OF ACTIVITY Each year, thousands of clinicians, basic scientists and other industry professionals sojourn to major international oncology conferences, like the American Society of Hematology (ASH) annual meeting, to hone their skills, network with colleagues and learn about recent advances altering state-of-the-art management in hematologic oncology. These events have become global stages where exciting science, cutting-edge concepts and practice-changing data emerge on a truly grand scale. This massive outpouring of information has enormous benefits for the hematologic oncology community, but the truth is it also creates a major challenge for practicing oncologists and hematologists. Although original data are consistently being presented and published, the flood of information unveiled during a major academic conference is unmatched and leaves in its wake an enormous volume of new knowledge that practicing oncologists must try to sift through, evaluate and consider applying. Unfortunately and quite commonly, time constraints and an inability to access these data sets leave many oncologists struggling to ensure that they’re aware of crucial practice-altering findings. This creates an almost insurmountable obstacle for clinicians in community practice because they are not only confronted almost overnight with thousands of new presentations and
    [Show full text]
  • Disruption of CSF-1R Signaling Inhibits Growth of AML with Inv(16)
    Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.uzh.ch Year: 2021 Disruption of CSF-1R signaling inhibits growth of AML with inv(16) Simonis, Alexander ; Russkamp, Norman F ; Mueller, Jan ; Wilk, C Matthias ; Wildschut, Mattheus H E ; Myburgh, Renier ; Wildner-Verhey van Wijk, Nicole ; Mueller, Rouven ; Balabanov, Stefan ; Valk, Peter J M ; Theocharides, Alexandre P A ; Manz, Markus G DOI: https://doi.org/10.1182/bloodadvances.2020003125 Posted at the Zurich Open Repository and Archive, University of Zurich ZORA URL: https://doi.org/10.5167/uzh-202789 Journal Article Published Version The following work is licensed under a Publisher License. Originally published at: Simonis, Alexander; Russkamp, Norman F; Mueller, Jan; Wilk, C Matthias; Wildschut, Mattheus H E; Myburgh, Renier; Wildner-Verhey van Wijk, Nicole; Mueller, Rouven; Balabanov, Stefan; Valk, Peter J M; Theocharides, Alexandre P A; Manz, Markus G (2021). Disruption of CSF-1R signaling inhibits growth of AML with inv(16). Blood advances, 5(5):1273-1277. DOI: https://doi.org/10.1182/bloodadvances.2020003125 STIMULUS REPORT Disruption of CSF-1R signaling inhibits growth of AML with inv(16) Alexander Simonis,1,* Norman F. Russkamp,1,* Jan Mueller,1 C. Matthias Wilk,1 Mattheus H. E. Wildschut,1,2 Renier Myburgh,1 Nicole Wildner-Verhey van Wijk,1 Rouven Mueller,1 Stefan Balabanov,1 Peter J. M. Valk,3 Alexandre P. A. Theocharides,1 and Markus G. Manz1 1Department of Medical Oncology and Hematology, University Hospital
    [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]
  • Pacritinib Combined with Sirolimus and Low-Dose Tacrolimus for GVHD
    Author Manuscript Published OnlineFirst on March 22, 2021; DOI: 10.1158/1078-0432.CCR-20-4725 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Pidala J et al GVHD prevention with combined JAK2/mTOR inhibition Pacritinib combined with sirolimus and low-dose tacrolimus for GVHD prevention after allogeneic hematopoietic cell transplantation: Preclinical and Phase I trial results Authors: Joseph Pidala1-3, Kelly Walton4, Hany Elmariah1,3, Jongphil Kim5, Asmita Mishra1,3, Nelli Bejanyan1,3, Taiga Nishihori1,3, Farhad Khimani1,3, Lia Perez1,3, Rawan G. Faramand1,3, Marco L. Davila1-3, Michael L. Nieder1,3, Elizabeth M. Sagatys6, Shernan G. Holtan4, Nicholas J. Lawrence7, Harshani R. Lawrence7, Bruce R. Blazar8, Claudio Anasetti1-3, Said M. Sebti9, Brian C. Betts4* Affiliations: 1Department of Blood and Marrow Transplantation – Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA, 2Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA, 3Department of Oncologic Sciences, University of South Florida, Tampa, FL, USA, 4Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA, 5Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA, 6Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, USA, 7Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL, USA, 8Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA, 9Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia USA. Running Title: GVHD prevention with combined JAK2/mTOR inhibition *Address correspondence to: Brian C. Betts, MD Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota Nils Hasselmo Hall, Room 2-108, 312 Church Street SE Minneapolis, MN 55455 [email protected] COI statement: B.C.B.
    [Show full text]
  • Comparison of Janus Kinase Inhibitors to Block the Type I Interferon Pathway in Human
    bioRxiv preprint doi: https://doi.org/10.1101/2021.02.08.430317; this version posted February 8, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Title: Comparison of Janus kinase inhibitors to block the type I interferon pathway in human skeletal muscle cells Authors: Travis B. Kinder1* and James Inglese1,2 1National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA, 2National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA *corresponding author: Travis B. Kinder, PhD Post-Doctoral Research Fellow, Assay Development and Screening Technology, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD, 20850 [email protected] Funding: This work was supported by a grant from the Cure Juvenile Myositis Foundation and the intramural program of NCATS, NIH, project 1ZIATR000342 (J.I.). Competing interests: None ORCID iD: Travis Kinder: 0000-0003-4161-8213 and James Inglese: 0000-0002-7332-5717 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.08.430317; this version posted February 8, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Abstract The family of Janus kinases (JAK1, JAK2, JAK3, TYK2) mediate signal transduction from cytokine receptors by phosphorylation and activation of intracellular signaling pathways and transcription factors.
    [Show full text]
  • Recommendations from York and Scarborough Medicines
    Recommendations from York and Scarborough Medicines Commissioning Committee July 2018 Drug name Indication Recommendation, rationale and place in RAG status Potential full year cost impact therapy CCG commissioned Technology Appraisals 1. Nil NHSE commissioned Technology Appraisals – for noting 2. TA520: Atezolizumab for Atezolizumab is recommended as an option for Red No cost impact to CCGs as NHS England treating locally advanced or treating locally advanced or metastatic non- commissioned. metastatic non-small-cell lung small-cell lung cancer (NSCLC) in adults who cancer after chemotherapy have had chemotherapy (and targeted treatment if they have an EGFR- or ALK‑ positive tumour), only if: atezolizumab is stopped at 2 years of uninterrupted treatment or earlier if the disease progresses and the company provides atezolizumab with the discount agreed in the patient access scheme. 3. TA522: Pembrolizumab for Pembrolizumab is recommended for use within Red No cost impact to CCGs as NHS England untreated locally advanced or the Cancer Drugs Fund as an option for commissioned. metastatic urothelial cancer untreated locally advanced or metastatic when cisplatin is unsuitable urothelial carcinoma in adults when cisplatin- containing chemotherapy is unsuitable, only if: pembrolizumab is stopped at 2 years of uninterrupted treatment or earlier if the disease progresses and the conditions of the managed access agreement for pembrolizumab are followed TA523: Midostaurin for Midostaurin is recommended, within its Red No cost impact to CCGs as NHS England untreated acute myeloid marketing authorisation, as an option in adults commissioned. leukaemia for treating newly diagnosed acute FLT3- mutation-positive myeloid leukaemia with standard daunorubicin and cytarabine as induction therapy, with high-dose cytarabine as consolidation therapy, and alone after complete response as maintenance therapy.
    [Show full text]
  • Annexes to the Annual Report of the European Medicines Agency 2014
    Annexes to the annual report of the European Medicines Agency 2014 Table of contents Annex 1 – Members of the Management Board ............................................................................. 2 Annex 2 – Members of the Committee for Medicinal Products for Human Use ................................... 4 Annex 3 – Members of the Pharmacovigilance Risk Assessment Committee ...................................... 6 Annex 4 – Members of the Committee for Medicinal Products for Veterinary Use ............................... 8 Annex 5 – Members of the Committee on Orphan Medicinal Products ............................................ 10 Annex 6 – Members of the Committee on Herbal Medicinal Products .............................................. 12 Annex 07 – Committee for Advanced Therapies .......................................................................... 14 Annex 8 – Members of the Paediatric Committee ........................................................................ 16 Annex 9 – Working parties and working groups .......................................................................... 18 Annex 10 – CHMP opinions in 2014 on medicinal products for human use ...................................... 22 Annex 11 – CVMP opinions in 2014 on medicinal products for veterinary use .................................. 36 Annex 12 – COMP opinions in 2014 on designation of orphan medicinal products ............................ 41 Annex 13 – HMPC European Union herbal monographs in 2014....................................................
    [Show full text]