EHA Library - The official digital education library of European Hematology Association (EHA)

METABOLIC ADAPTATIONS TO TARGETED THERAPY IN FLT3 MUTATED ACUTE MYELOID LEUKAEMIA (AML)
Author(s): ,
Paolo Gallipoli
Affiliations:
Haematology,UNIVERSITY OF CAMBRIDGE,Cambridge,United Kingdom
,
Ana SH Costa
Affiliations:
MRC Cancer Unit,UNIVERSITY OF CAMBRIDGE,Cambridge,United Kingdom
,
Konstantinos Tzelepis
Affiliations:
Wellcome Trust/Sanger Institute,UNIVERSITY OF CAMBRIDGE,Cambridge,United Kingdom
,
George Giotopoulos
Affiliations:
Haematology,UNIVERSITY OF CAMBRIDGE,Cambridge,United Kingdom
,
Shabana Vohra
Affiliations:
Haematology,UNIVERSITY OF CAMBRIDGE,Cambridge,United Kingdom
,
George Vassiliou
Affiliations:
Haematology,UNIVERSITY OF CAMBRIDGE,Cambridge,United Kingdom
,
Christian Frezza
Affiliations:
MRC Cancer Unit,UNIVERSITY OF CAMBRIDGE,Cambridge,United Kingdom
Brian Huntly
Affiliations:
Haematology,UNIVERSITY OF CAMBRIDGE,Cambridge,United Kingdom
(Abstract release date: 05/18/17) EHA Library. Gallipoli P. 06/23/17; 181423; S136
Paolo Gallipoli
Paolo Gallipoli
Contributions
Abstract

Abstract: S136

Type: Oral Presentation

Presentation during EHA22: On Friday, June 23, 2017 from 12:30 - 12:45

Location: Room N103

Background

FLT3 tyrosine kinase (TK) activating mutations (FLT-3mut) are amongst the most frequent in AML and are associated with a poor outcome. FLT-3mut promote constitutive activation of survival/proliferation pathways and have also been shown to lead to changes in cellular metabolism, such as increased glycolysis.
The FLT3 TK represents a valid therapeutic target and several FLT3 TK inhibitors (TKI) have been developed. However despite showing activity in the preclinical setting, FLT3 TKI have displayed limited efficacy in clinical trials. Resistance mechanisms to FLT3 TKI include receptor mutations and cell intrinsic adaptive mechanisms. Amongst the latter, metabolic adaptations might play a significant role although the exact mechanisms are still ill-defined.

Aims

We hypothesised that metabolic adaptations facilitate FLT3 TKI resistance and aimed to identify early metabolic changes in FLT-3mut AML, following TKI treatment, in an attempt to unveil novel therapeutic vulnerabilities. 

Methods

Liquid chromatography coupled to mass spectrometry (LC/MS), using stable isotope-based carbon flux tracing, and oxygen consumption rate/extracellular acidification rate as measured by an extracellular flux analyser (Seahorse, Agilent Technologies) were used to assess metabolic changes in FLT3mut cells after FLT3 TKI treatment. Gene expression changes were measured in the same conditions by RNA sequencing. Changes in viability and reactive oxygen species (ROS) in various culture conditions were measured by FACS. Gene silencing was performed using CRISPR-Cas9 gene editing and inducible short hairpin RNA interference.

Results

Analysis of published gene expression datasets demonstrated that glycolytic, citric acid cycle (CAC), and oxidative phosphorylation genes are upregulated in FLT-3mut compared to FLT3 wild-type (FLT3wt) patient samples at diagnosis. We then confirmed that both human and murine FLT-3mut cells display increased glycolytic and respiratory capacity compared to FLT-3wt cells. Upon treatment with the highly selective FLT-3 TKI AC220 (quizartinib), currently used in a number of clinical trials, these metabolic phenotypes were partially reversed. However, whilst glucose uptake was reduced upon FLT3 TK inhibition, glutamine uptake was not affected. Metabolic flux analysis using [U-13C]glutamine demonstrated that glutamine, while providing carbons for the CAC, was primarily used to support production of the major intracellular antioxidant glutathione upon AC220 treatment. This antioxidant function is necessary because, as expected, FLT-3mut cells displayed a large increase in ROS levels following TKI treatment when grown in the absence of glutamine and these changes correlated with a significant reduction in viability in the same conditions. Glutamine dependency of FLT3mut cells upon FLT3 TKI treatment was independently validated via a CRISPR-Cas9 drop-out genome-wide screen as glutaminase (GLS), the first enzyme in  glutamine catabolism, and several CAC enzymes were shown to be synthetically lethal with AC220. We went on to show that the combination of AC220 with a specific clinical grade GLS inhibitor (CB-839) or GLS gene silencing resulted in a significant reduction in viability and increase in ROS levels which could be rescued by supplementation of the media with the antioxidant N-acetylcysteine or a cell-permeable form of the CAC intermediate α-ketoglutarate.

Conclusion

Our data suggest that upon AC220 treatment, glutamine metabolism becomes a critical metabolic dependency in FLT3mut AML. Glutamine metabolism is mostly channelled towards glutathione production, while also supporting the CAC and both these fates contribute to its protective effects following FLT3 TK inhibition by respectively counteracting oxidative damage and sustaining macromolecule biosynthesis and cellular energetics. These data predict that a combined inhibition of glutamine metabolism and FLT3 TK activity may improve the eradication of FLT3mut AML cells.

Session topic: 3. Acute myeloid leukemia - Biology

Keyword(s): Targeted therapy, FLT3, Acute Myeloid Leukemia

Abstract: S136

Type: Oral Presentation

Presentation during EHA22: On Friday, June 23, 2017 from 12:30 - 12:45

Location: Room N103

Background

FLT3 tyrosine kinase (TK) activating mutations (FLT-3mut) are amongst the most frequent in AML and are associated with a poor outcome. FLT-3mut promote constitutive activation of survival/proliferation pathways and have also been shown to lead to changes in cellular metabolism, such as increased glycolysis.
The FLT3 TK represents a valid therapeutic target and several FLT3 TK inhibitors (TKI) have been developed. However despite showing activity in the preclinical setting, FLT3 TKI have displayed limited efficacy in clinical trials. Resistance mechanisms to FLT3 TKI include receptor mutations and cell intrinsic adaptive mechanisms. Amongst the latter, metabolic adaptations might play a significant role although the exact mechanisms are still ill-defined.

Aims

We hypothesised that metabolic adaptations facilitate FLT3 TKI resistance and aimed to identify early metabolic changes in FLT-3mut AML, following TKI treatment, in an attempt to unveil novel therapeutic vulnerabilities. 

Methods

Liquid chromatography coupled to mass spectrometry (LC/MS), using stable isotope-based carbon flux tracing, and oxygen consumption rate/extracellular acidification rate as measured by an extracellular flux analyser (Seahorse, Agilent Technologies) were used to assess metabolic changes in FLT3mut cells after FLT3 TKI treatment. Gene expression changes were measured in the same conditions by RNA sequencing. Changes in viability and reactive oxygen species (ROS) in various culture conditions were measured by FACS. Gene silencing was performed using CRISPR-Cas9 gene editing and inducible short hairpin RNA interference.

Results

Analysis of published gene expression datasets demonstrated that glycolytic, citric acid cycle (CAC), and oxidative phosphorylation genes are upregulated in FLT-3mut compared to FLT3 wild-type (FLT3wt) patient samples at diagnosis. We then confirmed that both human and murine FLT-3mut cells display increased glycolytic and respiratory capacity compared to FLT-3wt cells. Upon treatment with the highly selective FLT-3 TKI AC220 (quizartinib), currently used in a number of clinical trials, these metabolic phenotypes were partially reversed. However, whilst glucose uptake was reduced upon FLT3 TK inhibition, glutamine uptake was not affected. Metabolic flux analysis using [U-13C]glutamine demonstrated that glutamine, while providing carbons for the CAC, was primarily used to support production of the major intracellular antioxidant glutathione upon AC220 treatment. This antioxidant function is necessary because, as expected, FLT-3mut cells displayed a large increase in ROS levels following TKI treatment when grown in the absence of glutamine and these changes correlated with a significant reduction in viability in the same conditions. Glutamine dependency of FLT3mut cells upon FLT3 TKI treatment was independently validated via a CRISPR-Cas9 drop-out genome-wide screen as glutaminase (GLS), the first enzyme in  glutamine catabolism, and several CAC enzymes were shown to be synthetically lethal with AC220. We went on to show that the combination of AC220 with a specific clinical grade GLS inhibitor (CB-839) or GLS gene silencing resulted in a significant reduction in viability and increase in ROS levels which could be rescued by supplementation of the media with the antioxidant N-acetylcysteine or a cell-permeable form of the CAC intermediate α-ketoglutarate.

Conclusion

Our data suggest that upon AC220 treatment, glutamine metabolism becomes a critical metabolic dependency in FLT3mut AML. Glutamine metabolism is mostly channelled towards glutathione production, while also supporting the CAC and both these fates contribute to its protective effects following FLT3 TK inhibition by respectively counteracting oxidative damage and sustaining macromolecule biosynthesis and cellular energetics. These data predict that a combined inhibition of glutamine metabolism and FLT3 TK activity may improve the eradication of FLT3mut AML cells.

Session topic: 3. Acute myeloid leukemia - Biology

Keyword(s): Targeted therapy, FLT3, Acute Myeloid Leukemia

By clicking “Accept Terms & all Cookies” or by continuing to browse, you agree to the storing of third-party cookies on your device to enhance your user experience and agree to the user terms and conditions of this learning management system (LMS).

Cookie Settings
Accept Terms & all Cookies