Diagnostic value of 2-hydroxyglutarate detection by 1H MR spectroscopy in patients with glioma
Francesca Branzoli1,2, Anna Luisa Di Stefano2,3,4, Malgorzata Marjanska5, Romain Valabregue1,2, Stephane Lehericy1,2, and Marc Sanson2,3

1Brain and Spine Institute (ICM), Center for Neuroimaging Research (CENIR), F-75013, Paris, France, 2INSERM U1127/CNRS UMR7225, Sorbonne Universités, UPMC Univ Paris 06, ICM, F-75013, Paris, France, 3AP-HP, GH Pitié-Salpêtrière, Service de Neurologie 2, F-75013, Paris, France, 4Division of Neurology, Foch Hospital, Paris, France, 5Center for Magnetic Resonance Research and Department of Radiology, University of Minnesota, Minneapolis, MN, United States

Synopsis

Reliable quantification by magnetic resonance spectroscopy of the oncometabolite 2-hydroxyglutarate (2HG) has important implications in diagnosis of IDH mutation, prognosis, as well as assessment of the efficacy of anti-IDH targeted therapies. In this study, we employed two approaches for 2HG detection previously described, e.g., difference spectroscopy and optimized for 2HG detection conventional spectroscopy, in order to assess for the first time the specificity and sensitivity of the two methods, and to relate these results to the natural history and the neuroradiological status of patients with glioma.

Purpose

The overproduction of the oncometabolite 2-hydroxyglutarate (2HG) in IDH mutated gliomas [1, 2] can be detected non-invasively by magnetic resonance spectroscopy (MRS) [3-5]. Reliable quantification of 2HG has important implications in diagnosis of IDH mutation, prognosis, as well as assessment of the efficacy of anti-IDH targeted therapies. 2HG detection is very challenging and advanced methods for robust detection of this metabolite are not available in clinical settings. In this study, we employed two approaches for 2HG detection previously described, e.g., difference spectroscopy and optimized for 2HG detection conventional spectroscopy [3, 4], on a pre-operative cohort of patients, and on a post-operative cohort of patients previously treated with radio-and/or chemotherapy. The aim was to assess for the first time the specificity and sensitivity of the two methods, and to relate the results to the natural history and neuroradiological status of patients with glioma.

Materials and Methods

MRI/MRS protocol: Acquisitions were performed using a 3-T whole-body system (MAGNETOM Verio, Siemens, Erlangen, Germany) equipped with a 32-channel receive-only head coil. The protocol included T2-w FLAIR and T1-w sequences for voxel placement. MRS data were acquired using two sequences optimized for 2HG detection: 1) a single-voxel MEGA-PRESS [6] sequence (TR=2s, TE=68ms, 128 averages, scan time=9 min) with editing pulses applied at 1.9 and 7.5 ppm, for the edited and non-edited condition respectively, which allows to measure the 2HG signal at 4.05 ppm; 2) a single-voxel PRESS sequence (TR=2.5s, TE=97ms, TE1=32ms, TE2=65ms, 128 averages, scan time = 6 min), optimized to detect the 2HG signal at 2.25 ppm [3]. Water suppression was performed using VAPOR and outer volume suppression techniques [7]. Frequency and phase corrections were performed on single scans using the total choline signal at 3.2 ppm. Typical VOI size was 2x2x2 cm3 (Fig. 1c, f). VOI size was adapted to tumor size in order to minimize partial volume effects, keeping a minimum size of 6 cm3. Spectral quantification was performed using LCModel. The Cramer Rao lower bounds (CRLB) threshold for reliable 2HG detection was set to 20%.

Patient population: Thirty patients were included in the study: six subjects before surgery with suspected brain glioma (pre-operative cohort), and twenty-four patients who had surgery and radiotherapy or chemotherapy, and were affected by an IDH1 mutated glioma (post-operative cohort). In the post-operative cohort, five patients were affected by grade II, fourteen by grade III and five by grade IV IDH mutated glioma. In the same cohort, eleven patients were under radiotherapy or chemotherapy. IDH status was assessed for all patients combining detection of expression of IDH1-R132H mutant by immunohistochemistry (IHC) and Sanger sequencing for IDH1 and IDH2 gene mutations.

Results

In the pre-operative cohort, 2HG was detected in five out of six patients using MEGA-PRESS, while CRLB was < 20% only for two patients from PRESS data (examples of spectra in Fig. 1). Histopathological diagnosis after surgery confirmed the presence of IDH mutated gliomas in the five patients with detectable 2HG levels (two grade II and three grade III gliomas), while it revealed an IDH wild-type ganglioglioma in the patient with no 2HG signature [sensitivity 100% (0.4 to 1.0, 95% CI); specificity 100% (0.02 to 1.0, 95% CI)] (Table 1). In the post-operative cohort, 2HG was detected in seven out of twenty-four patients using MEGA-PRESS (sensitivity 29%), and only in three patients using PRESS (sensitivity 12%, data not shown). Detection of 2HG was not associated with grade (p=0.4), histological diagnosis (p=0.5), or the fact that patients were out of treatment at the moment of MRS examination (p=0.2).

Discussion and Conclusion

2HG spectroscopy is a promising technique for the diagnostic set-up and follow-up of glioma patients, which remains underused because of technical challenges. Preliminary results strongly suggest that difference spectroscopy (MEGA-PRESS) can provide high sensitivity/specificity for prediction of IDH mutation status before surgery, in contrast to optimized for 2HG detection conventional spectroscopy (PRESS). Fitting of conventional spectra can provide false-negative readouts due to spectral overlap of 2HG with chemically similar metabolites, such as glutamate, glutamine and GABA. The detection of 2HG is crucial for monitoring the response to treatments targeting IDH mutation. Although difference spectroscopy provides much higher sensitivity with respect to conventional MRS, in the post-operative cohort it was not possible to measure reliably 2HG concentration in 70% of patients. Some of the factors which can impact the sensitivity of the measurement are the residual tumor volume, 2HG concentration, cellular density, treatment, evolution of the disease, MRI phenotype, tumor localization, type of mutation and molecular pattern. All these elements together will be evaluated to provide a general guideline for 2HG detection after surgery.

Acknowledgements

Grant ‘Institut des neurosciences translationnelle - ANR-10-IAIHU-06', and 'Infrastructure d’avenir en Biologie Santé - ANR-11-INBS-0006’; P41 EB015894, and P30 NS076408 (MM).

References

[1] Parsons DW et al. Science 321:1807-1812 (2008). [2] Dang L et al. Nature. 462:739-744 (2009). [3] Choi C et al, Nature Med 18:624-29 (2012); [4] Andronesi OC et al, Science Transl Med 4:116ra4 (2012); [5] Pope WB et al, J Neuroonc. 107:197-205 (2012); [6] Mescher M et al. J Magn Reson, 123:226–229 (1996). [7] Tkac I et al, Magn Reson Med, 41:649–656 (1999).

Figures

Figure 1: MR spectra measured with MEGA-PRESS and PRESS in a patient with mutation IDH1 R132H (a, b) and in a patient with wild-type IDH glioma (d, e). VOI placement is shown for the two patients on an axial view of a T2-w FLAIR image (c and f, respectively).

Table 1: Correlation between 2HG detection by MEGA-PRESS and optimized PRESS, IDH mutation, and histological diagnosis, in the pre-operative cohort.



Proc. Intl. Soc. Mag. Reson. Med. 24 (2016)
4184