Your browser doesn't support javascript.
loading
Cotargeting of XPO1 Enhances the Antileukemic Activity of Midostaurin and Gilteritinib in Acute Myeloid Leukemia.
Brinton, Lindsey T; Sher, Steven; Williams, Katie; Canfield, Daniel; Orwick, Shelley; Wasmuth, Ronni; Cempre, Casey; Skinner, Jordan; Lehman, Amy; Blachly, James S; Byrd, John C; Lapalombella, Rosa.
Afiliação
  • Brinton LT; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Sher S; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Williams K; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Canfield D; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Orwick S; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Wasmuth R; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Cempre C; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Skinner J; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Lehman A; Center for Biostatistics, The Ohio State University, Columbus, OH 43210, USA.
  • Blachly JS; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
  • Byrd JC; Leukemia Research Program, The Ohio State University James Comprehensive Cancer Center, Columbus, OH 43210, USA.
  • Lapalombella R; Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA.
Cancers (Basel) ; 12(6)2020 Jun 14.
Article em En | MEDLINE | ID: mdl-32545904
ABSTRACT
Acute myeloid leukemia (AML) is a hematopoietic stem-cell-derived leukemia with often successive derived driver mutations. Late onset acquisition of internal tandem duplication in FLT3 (FLT3-ITD) at a high variant allele frequency often contributes to full transformation to a highly proliferative, rapidly progressive disease with poor outcome. The FLT3-ITD mutation is targetable with approved FLT3 small molecule inhibitors, including midostaurin and gilteritinib. However, outside of patients receiving allogeneic transplant, most patients fail to respond or relapse, suggesting alternative approaches of therapy will be required. We employed genome-wide pooled CRISPR knockout screening as a method for large-scale identification of targets whose knockout produces a phenotypic effect that enhances the antitumor properties of FLT3 inhibitors. Among the candidate targets we identified the effect of XPO1 knockout to be synergistic with midostaurin treatment. Next, we validated the genetic finding with pharmacologic combination of the slowly reversible XPO1 inhibitor selinexor with midostaurin and gilteritinib in FLT3-ITD AML cell lines and primary patient samples. Lastly, we demonstrated improved survival with either combination therapy compared to its monotherapy components in an aggressive AML murine model, supporting further evaluation and rapid clinical translation of this combination strategy.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2020 Tipo de documento: Article