4.7 Article

Glutaminolysis is a metabolic dependency in FLT3ITD acute myeloid leukemia unmasked by FLT3 tyrosine kinase inhibition

期刊

BLOOD
卷 131, 期 15, 页码 1639-1653

出版社

AMER SOC HEMATOLOGY
DOI: 10.1182/blood-2017-12-820035

关键词

-

资金

  1. Wellcome Trust [109967/Z/15/Z]
  2. Academy of Medical Sciences
  3. Lady Tata Memorial Trust
  4. European Research Council
  5. Medical Research Council
  6. Bloodwise
  7. Kay Kendall Leukaemia Fund
  8. Cambridge National Institute for Health Research Biomedical Research Centre
  9. Cancer Research UK [C56179/A21617]
  10. Cambridge Institute for Medical Research Flow Cytometry Core Facility
  11. Wellcome Trust [109967/Z/15/Z] Funding Source: Wellcome Trust
  12. Academy of Medical Sciences (AMS) [AMS-SGCL10-Gallipoli] Funding Source: researchfish
  13. Cancer Research UK [23015, 25508] Funding Source: researchfish
  14. European Hematology Association [TRTH157] Funding Source: researchfish
  15. Medical Research Council [MC_PC_12009, MR/M010392/1, MR/R009708/1, MC_UU_12022/6, G1000288] Funding Source: researchfish
  16. Wellcome Trust [109967/Z/15/Z] Funding Source: researchfish
  17. Worldwide Cancer Research [14-1069] Funding Source: researchfish
  18. MRC [MR/R009708/1, MC_UU_12022/6, G1000288, MR/M010392/1] Funding Source: UKRI

向作者/读者索取更多资源

FLT3 internal tandem duplication (FLT3(ITD)) mutations are common in acute myeloid leukemia (AML) associated with poor patient prognosis. Although new-generation FLT3 tyrosine kinase inhibitors (TKI) have shown promising results, the outcome of FLT3(ITD) AML patients remains poor and demands the identification of novel, specific, and validated therapeutic targets for this highly aggressive AML subtype. Utilizing an unbiased genomewide clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screen, we identify GLS, the first enzyme in glutamine metabolism, as synthetically lethal with FLT3-TKI treatment. Using complementary metabolomic and gene-expression analysis, we demonstrate that glutamine metabolism, through its ability to support both mitochondrial function and cellular redox metabolism, becomes a metabolic dependency of FLT3(ITD) AML, specifically unmasked by FLT3-TKI treatment. We extend thesefindings to AML subtypes driven by other tyrosine kinase (TK) activating mutations and validate the role of GLS as a clinically actionable therapeutic target in both primary AML and in vivo models. Our work highlights the role of metabolic adaptations as a resistance mechanism to several TKI and suggests glutaminolysis as a therapeutically targetable vulnerability when combined with specific TKI in FLT3(ITD) and other TK activating mutation-driven leukemias.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据