Modeling Oncogenic KIT Signaling and Drug Resistance in the Mouse

Lade...
Vorschaubild
Dateien
Dissertation Benedikt Bosbach OA.pdf
Dissertation Benedikt Bosbach OA.pdfGröße: 13.54 MBDownloads: 942
Datum
2012
Herausgeber:innen
Kontakt
ISSN der Zeitschrift
Electronic ISSN
ISBN
Bibliografische Daten
Verlag
Schriftenreihe
Auflagebezeichnung
DOI (zitierfähiger Link)
ArXiv-ID
Internationale Patentnummer
Angaben zur Forschungsförderung
Projekt
Open Access-Veröffentlichung
Open Access Green
Sammlungen
Core Facility der Universität Konstanz
Gesperrt bis
Titel in einer weiteren Sprache
Forschungsvorhaben
Organisationseinheiten
Zeitschriftenheft
Publikationstyp
Dissertation
Publikationsstatus
Published
Erschienen in
Zusammenfassung

KIT is a receptor tyrosine kinase critical for gametogenesis, hematopoiesis, melanogenesis, and the pacemaker system of the gastrointestinal tract, the interstitial cells of Cajal (ICC). The majority of gastrointestinal stromal tumors (GIST) harbor an activating mutation in the KIT gene. Introduction of such gain-of-function mutations into the endogenous Kit locus of the mouse, e.g., the knock-in of KitV558Δ, has been shown previously to result in interstitial cell of Cajal (ICC) hyperplasia throughout the gastrointestinal tract and development of cecal GIST lesions with full penetrance. This experimentally demonstrated the central role mutant KIT plays in the pathogenesis of GIST. Aberrant signal transduction in GIST lesions of KitV558Δ/+ mice includes sustained activation of the PI3K/AKT/S6, MAPK, and STAT3 pathways, which is amenable to inhibition with the tyrosine kinase inhibitor (TKI) imatinib. Imatinib is the first-line treatment for advanced GIST and can be given adjuvantly after surgery; it improves recurrence-free survival but is not curative. This necessitates chronic imatinib treatment, eventually resulting in relapse of patients. At this point, second-site mutations in Kit can be detected in the majority of the imatinib-resistant tumor lesions. A prominent second-site mutation abrogating the inhibitory effect of most TKIs is the so-called gatekeeper mutation, which affects a critical, conserved threonine residue in the kinase pocket of multiple oncogenic RTKs. The homologous mutations in human and murine KIT are KITT670I/+ and KitT669I, respectively.

As a proof-of-concept and, more importantly, to provide a preclinical model for the development of salvage therapies, the first aim of this study was the generation of a mouse model of imatinib-resistant GIST. By targeted mutagenesis the activating mutation KitV558Δ and the gatekeeper mutation KitT669I were introduced simultaneously into the Kit locus. The derived KitV558Δ;T669I/+ mice are viable and developed ICC hyperplasia and cecal GIST lesions. While GIST lesions were smaller in KitV558Δ;T669I/+ mice in comparison with KitV558Δ/+ mice, ICC hyperplasia was more pronounced, with no apparent changes in oncogenic signal transduction in both tissues/mouse models. Importantly, KitV558Δ;T669I/+ GIST were resistant to intervention with imatinib and dasatinib at the biochemical as well as at the histological/cellular level. This is further remarkable as both these TKIs are known to inhibit wild-type KIT (Kit+), which is present in the tumors of these heterozygous mice and likely in the majority of tumors of patients. The resistance mediated by the gatekeeper mutation was overcome by treatments with sunitinib and sorafenib, providing a rationale for their utilization in gatekeeper-mutant GIST cases.

Unexpectedly, KitV558Δ;T669I/+ mice uniformly and within six weeks after birth developed a polycythemia vera-like microcytic erythrocytosis. Macrocytic anemia is one of the hallmark phenotypes of mice with Kit loss-of-function dominant white-spotting (W) mutations. Together, this indicates that the KitV558Δ;T669I mutation is a strong gain-of-function mutation. Concordantly, KitV558Δ;T669I/+ mice had increased mast cell hyperplasia in addition to the observed microcytic erythrocytosis and the increased ICC hyperplasia. Besides the hyperplastic ICC and GIST lesions, also burst-forming units erythroid (BFU-Es) obtained from bone marrow and spleen of KitV558Δ;T669I/+ mice were resistant to imatinib and susceptible to sunitinib. This indicates that the observed erythrocytosis is largely dependent on aberrant kinase activity. It remains to be studied if the addition of the KitT669I mutation to the KitV558Δ mutation introduces qualitative changes in KIT, understanding of which might guide the design of improved inhibitors for gatekeeper-mutant kinases.

The second aim of this study was to molecularly abrogate specific phosphorylation sites of the KitV558Δ kinase to elucidate the contribution of the respective downstream signaling cascades to GIST development. Therefore, in the context of the KitV558Δ mutation two well-characterized phosphorylation sites of KIT, the Src family binding site pY567 and the PI3K binding site pY719, were substituted individually with phenylalanine by targeting the corresponding codons in the mouse genome. KitV558Δ;Y567F/Y567F mice developed attenuated GIST and ICC hyperplasia, associated with diminished activation of the MAPK pathway. Remarkably, GIST of KitV558Δ;Y567F/Y567F mice showed tumor progression along similar cytogenetic pathways as human GIST, with recurrent hemizygous deletions of mouse chr12, the chromosome which is largely syntenic to human chr14, the most frequently lost chromosome in GIST.

No tumor development was observed in the KitV558Δ;Y719F/Y719F mice up to the oldest animals analyzed at 23 months and median survival was wild type-like. Of note, ICC and accordingly their progenitors, which are the presumed cells-of-origin of GIST, were unaffected in KitV558Δ;Y719F/Y719F mice. While other KIT-dependent processes, namely hematopoiesis and melanogenesis, also did not show any apparent defects in KitV558Δ;Y719F/Y719F mice, the known male sterility phenotype of KitY719F/Y719F mice was not rescued in KitV558Δ;Y719F/Y719F mice. This indicates that cell-specific and absolute pathway requirements can exist in RTK signaling, even in the context of a potent activating mutation.

Translation of these genetic findings by treating established GIST of KitV558Δ/+ mice with a pharmacologic inhibitor of pan-class I PI3Ks, XL147, resulted in pathway-specific inhibition of ribosomal protein S6 and reduction of tumor cell proliferation. As responses to XL147 in comparison with results obtained in previous studies with imatinib were not complete, combination therapy with a MAPK pathway inhibitor might be of benefit, as indicated by the attenuated tumor growth of KitV558Δ;Y567F/Y567F mice. Approaches like this, to elucidate and target multiple proximal downstream mediators of oncogenic signaling, in the future might offer treatment options to circumvent the emergence of resistance to targeted cancer therapy and to potentiate it altogether.

Zusammenfassung in einer weiteren Sprache
Fachgebiet (DDC)
570 Biowissenschaften, Biologie
Schlagwörter
gametogenesis, spermatogenesis, melanogenesis, hematopoiesis, hematopoietic stem cell, HSC, hematopoietic progenitor cell, BFU-E, CFU-E, erythropoiesis, macrocytic anemia, microcytic erythrocytosis, polycythemia vera, EPO, JAK2, splenectomy, mast cell, receptor tyrosine kinase, KIT, interstitial cell of Cajal, ICC, hyperplasia, soft-tissue sarcoma, Gastrointestinaler Stromatumor, gastrointestinal stromal tumor, familial GIST, gain-of-function, loss-of-function, site-directed mutagenesis, conditional knock-in mouse model, loxP, signal transduction, PI3K, aneuploidy, chr12, chr14, tyrosine kinase inhibitor, sorafenib tosylate, dasatinib salt, sunitinib malate, drug resistance, gatekeeper
Konferenz
Rezension
undefined / . - undefined, undefined
Zitieren
ISO 690BOSBACH, Benedikt, 2012. Modeling Oncogenic KIT Signaling and Drug Resistance in the Mouse [Dissertation]. Konstanz: University of Konstanz
BibTex
@phdthesis{Bosbach2012Model-20467,
  year={2012},
  title={Modeling Oncogenic KIT Signaling and Drug Resistance in the Mouse},
  author={Bosbach, Benedikt},
  note={Teilw. ersch. in: Proceedings of the National Academy of Sciences of the United States of America : PNAS ; 109 (2012), 34. - E2276-E2283. - doi: 10.1073/pnas.1115240109},
  address={Konstanz},
  school={Universität Konstanz}
}
RDF
<rdf:RDF
    xmlns:dcterms="http://purl.org/dc/terms/"
    xmlns:dc="http://purl.org/dc/elements/1.1/"
    xmlns:rdf="http://www.w3.org/1999/02/22-rdf-syntax-ns#"
    xmlns:bibo="http://purl.org/ontology/bibo/"
    xmlns:dspace="http://digital-repositories.org/ontologies/dspace/0.1.0#"
    xmlns:foaf="http://xmlns.com/foaf/0.1/"
    xmlns:void="http://rdfs.org/ns/void#"
    xmlns:xsd="http://www.w3.org/2001/XMLSchema#" > 
  <rdf:Description rdf:about="https://kops.uni-konstanz.de/server/rdf/resource/123456789/20467">
    <foaf:homepage rdf:resource="http://localhost:8080/"/>
    <dspace:hasBitstream rdf:resource="https://kops.uni-konstanz.de/bitstream/123456789/20467/1/Dissertation%20Benedikt%20Bosbach%20OA.pdf"/>
    <dspace:isPartOfCollection rdf:resource="https://kops.uni-konstanz.de/server/rdf/resource/123456789/28"/>
    <dc:rights>terms-of-use</dc:rights>
    <void:sparqlEndpoint rdf:resource="http://localhost/fuseki/dspace/sparql"/>
    <bibo:uri rdf:resource="http://kops.uni-konstanz.de/handle/123456789/20467"/>
    <dcterms:abstract xml:lang="eng">KIT is a receptor tyrosine kinase critical for gametogenesis, hematopoiesis, melanogenesis, and the pacemaker system of the gastrointestinal tract, the interstitial cells of Cajal (ICC). The majority of gastrointestinal stromal tumors (GIST) harbor an activating mutation in the KIT gene. Introduction of such gain-of-function mutations into the endogenous Kit locus of the mouse, e.g., the knock-in of Kit&lt;sup&gt;V558Δ&lt;/sup&gt;, has been shown previously to result in interstitial cell of Cajal (ICC) hyperplasia throughout the gastrointestinal tract and development of cecal GIST lesions with full penetrance. This experimentally demonstrated the central role mutant KIT plays in the pathogenesis of GIST. Aberrant signal transduction in GIST lesions of Kit&lt;sup&gt;V558Δ/+&lt;/sup&gt; mice includes sustained activation of the PI3K/AKT/S6, MAPK, and STAT3 pathways, which is amenable to inhibition with the tyrosine kinase inhibitor (TKI) imatinib. Imatinib is the first-line treatment for advanced GIST and can be given adjuvantly after surgery; it improves recurrence-free survival but is not curative. This necessitates chronic imatinib treatment, eventually resulting in relapse of patients. At this point, second-site mutations in Kit can be detected in the majority of the imatinib-resistant tumor lesions. A prominent second-site mutation abrogating the inhibitory effect of most TKIs is the so-called gatekeeper mutation, which affects a critical, conserved threonine residue in the kinase pocket of multiple oncogenic RTKs. The homologous mutations in human and murine KIT are KIT&lt;sup&gt;T670I/+&lt;/sup&gt; and Kit&lt;sup&gt;T669I&lt;/sup&gt;, respectively.&lt;br /&gt;&lt;br /&gt; As a proof-of-concept and, more importantly, to provide a preclinical model for the development of salvage therapies, the first aim of this study was the generation of a mouse model of imatinib-resistant GIST. By targeted mutagenesis the activating mutation Kit&lt;sup&gt;V558Δ&lt;/sup&gt; and the gatekeeper mutation Kit&lt;sup&gt;T669I&lt;/sup&gt; were introduced simultaneously into the Kit locus. The derived Kit&lt;sup&gt;V558Δ;T669I/+&lt;/sup&gt; mice are viable and developed ICC hyperplasia and cecal GIST lesions. While GIST lesions were smaller in Kit&lt;sup&gt;V558Δ;T669I/+&lt;/sup&gt; mice in comparison with Kit&lt;sup&gt;V558Δ/+&lt;/sup&gt; mice, ICC hyperplasia was more pronounced, with no apparent changes in oncogenic signal transduction in both tissues/mouse models. Importantly, Kit&lt;sup&gt;V558Δ;T669I/+&lt;/sup&gt; GIST were resistant to intervention with imatinib and dasatinib at the biochemical as well as at the histological/cellular level. This is further remarkable as both these TKIs are known to inhibit wild-type KIT (Kit&lt;sup&gt;+&lt;/sup&gt;), which is present in the tumors of these heterozygous mice and likely in the majority of tumors of patients. The resistance mediated by the gatekeeper mutation was overcome by treatments with sunitinib and sorafenib, providing a rationale for their utilization in gatekeeper-mutant GIST cases.&lt;br /&gt;&lt;br /&gt; Unexpectedly, Kit&lt;sup&gt;V558Δ;T669I/+&lt;/sup&gt; mice uniformly and within six weeks after birth developed a polycythemia vera-like microcytic erythrocytosis. Macrocytic anemia is one of the hallmark phenotypes of mice with Kit loss-of-function dominant white-spotting (W) mutations. Together, this indicates that the Kit&lt;sup&gt;V558Δ;T669I&lt;/sup&gt; mutation is a strong gain-of-function mutation. Concordantly, Kit&lt;sup&gt;V558Δ;T669I/+&lt;/sup&gt; mice had increased mast cell hyperplasia in addition to the observed microcytic erythrocytosis and the increased ICC hyperplasia. Besides the hyperplastic ICC and GIST lesions, also burst-forming units erythroid (BFU-Es) obtained from bone marrow and spleen of Kit&lt;sup&gt;V558Δ;T669I/+&lt;/sup&gt; mice were resistant to imatinib and susceptible to sunitinib. This indicates that the observed erythrocytosis is largely dependent on aberrant kinase activity. It remains to be studied if the addition of the Kit&lt;sup&gt;T669I&lt;/sup&gt; mutation to the Kit&lt;sup&gt;V558Δ&lt;/sup&gt; mutation introduces qualitative changes in KIT, understanding of which might guide the design of improved inhibitors for gatekeeper-mutant kinases.&lt;br /&gt;&lt;br /&gt; The second aim of this study was to molecularly abrogate specific phosphorylation sites of the Kit&lt;sup&gt;V558Δ&lt;/sup&gt; kinase to elucidate the contribution of the respective downstream signaling cascades to GIST development. Therefore, in the context of the Kit&lt;sup&gt;V558Δ&lt;/sup&gt; mutation two well-characterized phosphorylation sites of KIT, the Src family binding site pY567 and the PI3K binding site pY719, were substituted individually with phenylalanine by targeting the corresponding codons in the mouse genome. Kit&lt;sup&gt;V558Δ;Y567F/Y567F&lt;/sup&gt; mice developed attenuated GIST and ICC hyperplasia, associated with diminished activation of the MAPK pathway. Remarkably, GIST of Kit&lt;sup&gt;V558Δ;Y567F/Y567F&lt;/sup&gt; mice showed tumor progression along similar cytogenetic pathways as human GIST, with recurrent hemizygous deletions of mouse chr12, the chromosome which is largely syntenic to human chr14, the most frequently lost chromosome in GIST.&lt;br /&gt;&lt;br /&gt; No tumor development was observed in the Kit&lt;sup&gt;V558Δ;Y719F/Y719F&lt;/sup&gt; mice up to the oldest animals analyzed at 23 months and median survival was wild type-like. Of note, ICC and accordingly their progenitors, which are the presumed cells-of-origin of GIST, were unaffected in Kit&lt;sup&gt;V558Δ;Y719F/Y719F&lt;/sup&gt; mice. While other KIT-dependent processes, namely hematopoiesis and melanogenesis, also did not show any apparent defects in Kit&lt;sup&gt;V558Δ;Y719F/Y719F&lt;/sup&gt; mice, the known male sterility phenotype of Kit&lt;sup&gt;Y719F/Y719F&lt;/sup&gt; mice was not rescued in Kit&lt;sup&gt;V558Δ;Y719F/Y719F&lt;/sup&gt; mice. This indicates that cell-specific and absolute pathway requirements can exist in RTK signaling, even in the context of a potent activating mutation.&lt;br /&gt;&lt;br /&gt; Translation of these genetic findings by treating established GIST of Kit&lt;sup&gt;V558Δ/+&lt;/sup&gt; mice with a pharmacologic inhibitor of pan-class I PI3Ks, XL147, resulted in pathway-specific inhibition of ribosomal protein S6 and reduction of tumor cell proliferation. As responses to XL147 in comparison with results obtained in previous studies with imatinib were not complete, combination therapy with a MAPK pathway inhibitor might be of benefit, as indicated by the attenuated tumor growth of Kit&lt;sup&gt;V558Δ;Y567F/Y567F&lt;/sup&gt; mice. Approaches like this, to elucidate and target multiple proximal downstream mediators of oncogenic signaling, in the future might offer treatment options to circumvent the emergence of resistance to targeted cancer therapy and to potentiate it altogether.</dcterms:abstract>
    <dcterms:issued>2012</dcterms:issued>
    <dc:creator>Bosbach, Benedikt</dc:creator>
    <dc:contributor>Bosbach, Benedikt</dc:contributor>
    <dc:date rdf:datatype="http://www.w3.org/2001/XMLSchema#dateTime">2012-11-15T09:24:49Z</dc:date>
    <dc:language>eng</dc:language>
    <dcterms:title>Modeling Oncogenic KIT Signaling and Drug Resistance in the Mouse</dcterms:title>
    <dcterms:isPartOf rdf:resource="https://kops.uni-konstanz.de/server/rdf/resource/123456789/28"/>
    <dcterms:rights rdf:resource="https://rightsstatements.org/page/InC/1.0/"/>
    <dcterms:hasPart rdf:resource="https://kops.uni-konstanz.de/bitstream/123456789/20467/1/Dissertation%20Benedikt%20Bosbach%20OA.pdf"/>
  </rdf:Description>
</rdf:RDF>
Interner Vermerk
xmlui.Submission.submit.DescribeStep.inputForms.label.kops_note_fromSubmitter
Kontakt
URL der Originalveröffentl.
Prüfdatum der URL
Prüfungsdatum der Dissertation
September 12, 2012
Finanzierungsart
Kommentar zur Publikation
Teilw. ersch. in: Proceedings of the National Academy of Sciences of the United States of America : PNAS ; 109 (2012), 34. - E2276-E2283. - doi: 10.1073/pnas.1115240109
Allianzlizenz
Corresponding Authors der Uni Konstanz vorhanden
Internationale Co-Autor:innen
Universitätsbibliographie
Begutachtet
Diese Publikation teilen