Study of the Mutated Isocitrate Dehydrogenase 1 in Acute Myeloid Leukemia Using Hyperpolarized [1-13C]α-ketoglutaric Acid
Eugen Kubala1,2,3, Kim A. Muñoz Álvarez1, Oliver Dovey4, Steffen J. Glaser2, Markus Schwaiger1, George S. Vassiliou4, Roland Rad5,6, Rolf F. Schulte3, and Marion I. Menzel3

1Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany, 2Department of Chemistry, Technische Universität München, Munich, Germany, 3General Electric Global Research, Munich, Germany, 4The Welcome Trust Sanger Institute, Hinxton/Cambridge, United Kingdom, 5Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany, 6Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Munich, Germany

Synopsis

Previous studies suggest that isocitrate dehydrogenase 1 (IDH1) mutation plays a significant role in the cancerous metabolome. Among other alternations, expression of branched chain amino-acid transaminase 1 (BCAT1) is reduced, causing a decrease of α-ketoglutaric acid (αKG) to glutamic acid metabolic pathway. More importantly, the mutated IDH1 catalyzes a reaction of αKG to the oncometabolite 2-hydroxyglutarate. In this study we proved that these metabolic changes can be measured using hyperpolarized [1-13C]α-KG and 13Cmetabolic magnetic resonance spectroscopy (13CMMRS) in acute myeloid leukemia cell line in vitro.

Purpose

The aim was to study the influence of isocitrate dehydrogenase 1 mutation IDH1R132H on α-ketoglutarate metabolism in an acute myeloid leukemia cell line in vitro using hyperpolarized [1-13C]α-ketoglutarate and 13C metabolic magnetic resonance spectroscopy (13CMMRS).

Introduction

The isocitrate dehydrogenase 1 (IDH1) is an intracellular enzyme, located in the cytosol, which normally catalyzes the oxidation of isocitrate to α-ketoglutarate (αKG). However, in several cancer types, such as glioma or acute myeloid leukemia (AML), R132H mutation in the IDH1 gene causes a functional change, which plays a significant role in further cancer development1,2. The monoallelic mutation causes a gain of function, resulting in an alternative product from αKG’s oxidation; the oncometabolite 2-hydroxyglutarate (2-HG)3. This causes a 10–100 times higher accumulation of 2-HG in the cancerous than in healthy tissue4,5, allowing even its detection by 1H magnetic resonance spectroscopy6. Among other alternations caused by IDH1 mutation, expression of branched chain amino-acid transaminase 1 (BCAT1) is reduced causing decrease in αKG to glutamic acid (Glu) metabolic pathway7,8. See Fig.1 for detailed metabolic pathways description. Interestingly, this mutated version of the IDH1 gene, was discovered in 8% of all acute myeloid leukemia (AML) genomes tested and was largely connected with normal cytogenetic status; it was present in 16% cytogenetically normal samples9. Dissolution dynamic nuclear polarization (dDNP) of 13C-labeled compounds enhances the NMR signal in vivo by over 10,000 times, which enables studying metabolic pathways in real time10.

Methods

An acute myeloid leukemia (AML) cell line, carrying IDH1R132H, Npm1cA and NrasG12D was derived from mutant IDH1R132H (Cre recombined) mouse. As a control AML cell line carrying Npm1cA and NrasG12D only was used. Both cell lines were cultured in X-Vivo 20 medium (Lonza, Basel, Switzerland) supplemented with IL-3, IL-6, SCF (Peprotech, Rocky Hill, NJ, USA) and 5% FCS (Biochrom, Berlin, Germany) and grown on special low-attachment plates (Corning, Corning, NY, USA). Cells were kept in a humidified atmosphere at 5% CO2 and 37°C. Mixture of [1-13C]α-ketoglutaric acid (CIL, Massachusetts, USA), 20 mM trityl radical OX063 (GE Healthcare, Amersham, UK), and 0.6 mM Dotarem® (Guerbet, Roissy, France) in DMSO was hyperpolarized using Hypersense™ polarizer (Oxford Instruments, Abingdon, UK) at ~94 GHz. The sample was dissolved in PBS buffer to 20 mM at pH 7.4. The solution was mixed with AML-IDH1mut/IDH1wt cell suspension (5×107 cells/mL) resulting in 5 mM final [1-13C]α-ketoglutarate concentration. The mixture was transferred to 5mm NMR tube and measured at 1 Tesla Spinsolve® NMR spectrometer (Magritek, San Diego, USA) using FA = 10°, TR = 3 s, NS = 100.

Results & Discussion

We were able to observe a differences in the metabolism of [1-13C]α-KG caused by the IDH1R132H mutation using hyperpolarized [1-13C]α-KG and metabolic magnetic resonance spectroscopy (MMRS) in AML cell line in vitro. (Fig.2) The longitudinal (or spin-lattice) relaxation time (T1) allowed long enough measuring time to obtain the metabolism with T1 = 57 ± 3 s for [1-13C]α-KG and 56 ± 3 s for [1-13C]α-ketoglutarate hydrate at 1 Tesla in vitro (n=4). Our data proved that IDH1R132H mutation causes a significant decrease of αKG (172.6 ppm) to Glu (177.2 ppm) metabolism by 89 ± 8% (Fig.3). Therefore, we assume that expression of BCAT1 is reduced in AML as in previously reported gliomas.7,8 Due to a low spectral resolution of 1 Tesla spectrometer, we are not absolutely positive, whether the peak at 183.7 ppm belongs to oncometabolite 2-HG or to [5-13C]αKG, even though 99% [1-13C]αKG was used for the experiments.

Acknowledgements

Funded by DFG SFB824 Teilprojekt A5

References

1) Yang H, et al. Clin Cancer Res. 2012;18:5562–5571. 2) Losman JA, et al. Genes Dev. 2013;27:836–852. 3) Walker EJ, et al. Cancer research. 2012;72:636–644. 4) Ward PS, et al. Cancer Cell. 2010;17:225–234. 5) Dang L, et al. Nature. 2009;462:739–744. 6) Choi C, et al. Nat Med. 2012;18(4):624-629. 7) Tonjes M, et al. Nat Med. 2013;19:901-908. 8) Chaumeil MM, et al. Cancer Res. 2014;74(16):4247-4257 9) Mardis ER, et al. N Eng J Med. 2009;361:1058–1066. 10) Ardenkjær-Larsen JH, et al. Proc Natl Acad Sci U.S.A. 2003;100(18):10158

Figures

Fig.1: α-ketoglutarate metabolism. Isocitrate dehydrogenase 1 (IDH1) oxidates isocitrate to α-KG; mutated IDH1mut metabolizes α-KG into 2-HG. Branched chain amino-acid transaminase 1 (BCAT1) catalyzes the reversible (rev.) transamination of α-KG to Glu. Alanine transaminase 1 (ALT1) catalyzes rev. transamination of alanine to α-KG. Glutamate dehydrogenase (GDH1) catalyzes rev. reaction of α-KG to Glu. Aspartate transaminase 1 (AST) catalyzes reversible transamination between aspartate and Glu.

Fig.2: Spectra obtained showing the sum of first 60 s (TR = 3 s) obtained after adding hyperpolarized [1-13C]α-ketoglutarate (172.6 ppm) in (a) IDH1mut cell line (b) IDH1wt cell line, and (c) control buffer solution. The spectra show significant difference in [1-13C]glutamate (177.2 ppm). Other peaks: [1-13C]α-KG hydratate (180.4ppm), [5-13C]α-KG + [1-13C]2-HG (183.7 ppm), other peaks are most probably contamination.

Fig.3: Comparison of the signal intensity of [1-13C]glutamate normalized to sum of all hyperpolarized metabolites obtained in IDH1wt cell line (n=2) and IDH1mut cell line (n=2). The metabolism of α-KG to glutamate decreases by 89 ± 8% in IDH1mut compared to IDH1wt cell line.



Proc. Intl. Soc. Mag. Reson. Med. 24 (2016)
0891