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

IDH MUTATIONS DRIVE LEUKEMOGENESIS BY ONCOGENIC ACTIVATION DUE TO INSULATOR DYSFUNCTION
Author(s): ,
Sophie Steinhäuser
Affiliations:
Internal Medicine II (Hematology/Oncology),University Hospital Schleswig-Holstein,Kiel,Germany
,
Patricia Silva
Affiliations:
Department of Hematology and Oncology,Charité University Hospital,Berlin,Germany
,
Sonja Hänzelmann
Affiliations:
Internal Medicine II (Hematology/Oncology),University Hospital Schleswig-Holstein,Kiel,Germany
,
Martin Neumann
Affiliations:
Internal Medicine II (Hematology/Oncology),University Hospital Schleswig-Holstein,Kiel,Germany
,
Lorenz Bastian
Affiliations:
Internal Medicine II (Hematology/Oncology),University Hospital Schleswig-Holstein,Kiel,Germany
,
Kathrin Richter
Affiliations:
Internal Medicine II (Hematology/Oncology),University Hospital Schleswig-Holstein,Kiel,Germany
,
Shuli Xia
Affiliations:
Kennedy Krieger Institute,Baltimore, Maryland,United States;Department of Neurology,Johns Hopkins University,Baltimore, Maryland,United States
,
Lennart Lenk
Affiliations:
Department of Pediatrics I, ALL-BFM Study Group,University Hospital Schleswig-Holstein,Kiel,Germany
,
Fotini Vogiatzi
Affiliations:
Department of Pediatrics I, ALL-BFM Study Group,University Hospital Schleswig-Holstein,Kiel,Germany
,
Dennis M. Schewe
Affiliations:
Department of Pediatrics I, ALL-BFM Study Group,University Hospital Schleswig-Holstein,Kiel,Germany
,
Malte Spielmann
Affiliations:
Institute for Human Genetics,University Hospital Schleswig-Holstein,Lübeck,Germany
,
Christoph Röllig
Affiliations:
Internal Medicine I,University Hospital Carl-Gustav-Carus,Dresden,Germany
Claudia D. Baldus
Affiliations:
Internal Medicine II (Hematology/Oncology),University Hospital Schleswig-Holstein,Kiel,Germany
EHA Library. Steinhäuser S. 06/09/21; 325129; EP375
Sophie Steinhäuser
Sophie Steinhäuser
Contributions
Abstract
Presentation during EHA2021: All e-poster presentations will be made available as of Friday, June 11, 2021 (09:00 CEST) and will be accessible for on-demand viewing until August 15, 2021 on the Virtual Congress platform.

Abstract: EP375

Type: E-Poster Presentation

Session title: Acute myeloid leukemia - Biology & Translational Research

Background
Acute myeloid leukemia (AML) is a heterogenous disease, characterized by complex patterns of molecular alterations and somatic driver mutations in genes of numerous functional categories, such as NPM1, DNMT3A or IDH1/2. In younger adults, IDH1 mutations occur in 6-10% of AML cases, whereas elderly AML patients show a significantly higher IDH1 mutation frequency of 17.2% accompanied by DNA hypermethylation. Despite advances in targeted therapy due to the recent approval of IDH inhibitors (Enasidenib, Ivosidenib), elderly AML patients with IDH mutations still have an inferior outcome irrespective of cytogenetic group with high rates of chemotherapy resistance and relapses. Recently, the IDH1 p.R132H mutation was linked to altered DNA loop formation by disrupted CTCF binding, leading to oncogene activation in glioma cells, which correlated with inferior outcome. 

Aims

Here, we investigate the role of the IDH1 p.R132H mutation in altering 3D DNA architecture and subsequent activation of oncogene expression in AML.

Methods

To identify upregulated oncogenes in IDH1-mutant (-mut) AML, differential gene expression was analyzed in two independent AML datasets, the BeatAML RNA-Seq dataset with n = 264 patients with annotated IDH1-status (Tyner et al. 2018) and a microarray dataset with n = 247 patients (Verhaak et al. 2009). To validate enhanced oncogene expression by RT-qPCR and investigate DNA loop formation, we generated a KG1a-based cellular model containing the IDH1 p.R132H point mutation using CRISPR base editing. Confirmed oncogenes were correlated with differential occupancy and methylation of corresponding CTCF binding sites using chromatin immunoprecipitation (ChIP) and methylation-sensitive restriction followed by RT-qPCR. Clinical relevance of identified oncogenes was investigated by comparing survival of patients with high or low oncogene expression with or without IDH1 mutation in two independent clinical datasets (BeatAML and Bamopoulos et al. 2020, n = 246).

Results

Differential expression analysis of AML datasets identified 179 genes upregulated by >1.5 fold (ANOVA, p < 0.05) in IDH1-mut AML patients compared to IDH1-wildtype (-wt) patients, including 13 oncogenes. Upregulated oncogene expression was confirmed in the KG1a IDH1 p.R132H model revealing highest upregulation (2.5 fold) of the receptor tyrosine kinase gene PDGFRA. Quantification of CTCF occupancy identified decreased CTCF binding by 30% at the CTCF anchor region 1.7 Mb upstream of the PDGFRA locus. In addition, DNA methylation of the identified CTCF binding site was increased by 2-fold in cells harboring the IDH1 p.R132H mutation. Analysis of AML clinical data revealed significantly decreased survival of AML patients with high PDGFRA expression among IDH1-mut but not IDH1-wt patients (Fig. 1, OS of IDH1-mut/-wt patients with high/low PDGFRA, data: Bamopoulos). Interestingly, IDH1-mut KG1a cells showed higher sensitivity towards the tyrosine kinase inhibitor Dasatinib in a WST-based drug screen, supporting Dasatinib as novel candidate for treatment of AML patients with IDH1 mutation.

Conclusion

Our data suggest that IDH1 p.R132H mutation leads to 3D conformational changes in AML, disrupting DNA loop formation and insulation of the tyrosine kinase PDGFRA, which subsequently results in upregulated PDGFRA oncogene expression. As high PDGFRA expression correlated with poor prognosis, treatment with tyrosine kinase inhibitor Dasatinib may be an option for IDH1 p.R132H mutant AML patients. Our findings offer a base for novel treatment strategies and tackling resistance in AML patients with IDH1 mutations.

Keyword(s): AML, Chromatin structure, PDGFRA, Tyrosine kinase inhibitor

Presentation during EHA2021: All e-poster presentations will be made available as of Friday, June 11, 2021 (09:00 CEST) and will be accessible for on-demand viewing until August 15, 2021 on the Virtual Congress platform.

Abstract: EP375

Type: E-Poster Presentation

Session title: Acute myeloid leukemia - Biology & Translational Research

Background
Acute myeloid leukemia (AML) is a heterogenous disease, characterized by complex patterns of molecular alterations and somatic driver mutations in genes of numerous functional categories, such as NPM1, DNMT3A or IDH1/2. In younger adults, IDH1 mutations occur in 6-10% of AML cases, whereas elderly AML patients show a significantly higher IDH1 mutation frequency of 17.2% accompanied by DNA hypermethylation. Despite advances in targeted therapy due to the recent approval of IDH inhibitors (Enasidenib, Ivosidenib), elderly AML patients with IDH mutations still have an inferior outcome irrespective of cytogenetic group with high rates of chemotherapy resistance and relapses. Recently, the IDH1 p.R132H mutation was linked to altered DNA loop formation by disrupted CTCF binding, leading to oncogene activation in glioma cells, which correlated with inferior outcome. 

Aims

Here, we investigate the role of the IDH1 p.R132H mutation in altering 3D DNA architecture and subsequent activation of oncogene expression in AML.

Methods

To identify upregulated oncogenes in IDH1-mutant (-mut) AML, differential gene expression was analyzed in two independent AML datasets, the BeatAML RNA-Seq dataset with n = 264 patients with annotated IDH1-status (Tyner et al. 2018) and a microarray dataset with n = 247 patients (Verhaak et al. 2009). To validate enhanced oncogene expression by RT-qPCR and investigate DNA loop formation, we generated a KG1a-based cellular model containing the IDH1 p.R132H point mutation using CRISPR base editing. Confirmed oncogenes were correlated with differential occupancy and methylation of corresponding CTCF binding sites using chromatin immunoprecipitation (ChIP) and methylation-sensitive restriction followed by RT-qPCR. Clinical relevance of identified oncogenes was investigated by comparing survival of patients with high or low oncogene expression with or without IDH1 mutation in two independent clinical datasets (BeatAML and Bamopoulos et al. 2020, n = 246).

Results

Differential expression analysis of AML datasets identified 179 genes upregulated by >1.5 fold (ANOVA, p < 0.05) in IDH1-mut AML patients compared to IDH1-wildtype (-wt) patients, including 13 oncogenes. Upregulated oncogene expression was confirmed in the KG1a IDH1 p.R132H model revealing highest upregulation (2.5 fold) of the receptor tyrosine kinase gene PDGFRA. Quantification of CTCF occupancy identified decreased CTCF binding by 30% at the CTCF anchor region 1.7 Mb upstream of the PDGFRA locus. In addition, DNA methylation of the identified CTCF binding site was increased by 2-fold in cells harboring the IDH1 p.R132H mutation. Analysis of AML clinical data revealed significantly decreased survival of AML patients with high PDGFRA expression among IDH1-mut but not IDH1-wt patients (Fig. 1, OS of IDH1-mut/-wt patients with high/low PDGFRA, data: Bamopoulos). Interestingly, IDH1-mut KG1a cells showed higher sensitivity towards the tyrosine kinase inhibitor Dasatinib in a WST-based drug screen, supporting Dasatinib as novel candidate for treatment of AML patients with IDH1 mutation.

Conclusion

Our data suggest that IDH1 p.R132H mutation leads to 3D conformational changes in AML, disrupting DNA loop formation and insulation of the tyrosine kinase PDGFRA, which subsequently results in upregulated PDGFRA oncogene expression. As high PDGFRA expression correlated with poor prognosis, treatment with tyrosine kinase inhibitor Dasatinib may be an option for IDH1 p.R132H mutant AML patients. Our findings offer a base for novel treatment strategies and tackling resistance in AML patients with IDH1 mutations.

Keyword(s): AML, Chromatin structure, PDGFRA, Tyrosine kinase inhibitor

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