Abstract
Glioblastomas are aggressive brain tumors with dismal prognosis. One of the main bottlenecks for developing more effective therapies for glioblastoma stems from their histologic and molecular heterogeneity, leading to distinct tumor microenvironments and disease phenotypes. Effectively characterizing these features would improve the clinical management of glioblastoma. Glucose flux rates through glycolysis and mitochondrial oxidation have been recently shown to quantitatively depict glioblastoma proliferation in mouse models (GL261 and CT2A tumors, 38±3 mm3) using dynamic glucose-enhanced (DGE) deuterium spectroscopy. However, the spatial features of tumor microenvironment phenotypes remain hitherto unresolved. Here, we develop a DGE Deuterium Metabolic Imaging (DMI) approach for profiling tumor microenvironments through glucose conversion kinetics. Using a multimodal combination of tumor mouse models, novel strategies for spectroscopic imaging and noise attenuation, and histopathological correlations, we show that tumor lactate turnover mirrors phenotype differences between GL261 and CT2A mouse glioblastoma (59±7 mm3), whereas peritumoral glutamate-glutamine recycling is a potential marker of invasion capacity in pooled cohorts, linked to secondary brain lesions. Our findings were validated by histopathological characterization of each tumor, including cell density and proliferation, peritumoral infiltration, and distant migration. Our study bodes well for precision neuro-oncology, highlighting the importance of mapping glucose flux rates to better understand the metabolic heterogeneity of glioblastoma and its links to disease phenotypes.
Introduction
Glioblastoma (glioma grade 4 or GBM) are the most aggressive primary brain tumors in adults. The dismal prognosis of such heterogeneous tumors is mostly attributed to recurrence, associated with limited response to treatment and an infiltrative pattern that prevents full surgical resection [1]. Glioblastoma heterogeneity is reflected in the tumor microenvironment, where glioma cells constantly adapt to their evolving microhabitats, with different biophysical characteristics, progression stages, and therapy resistance [2]. To sustain active proliferation, cancer cells exchange metabolic intermediates with their microenvironment [3] and undergo metabolic reprogramming [4], relying heavily on aerobic glycolysis – upregulation of glucose uptake concomitant with lactate synthesis, leading to acidification of the tumor microenvironment. While this so-called Warburg effect [5] favors e.g. invasion [6], metabolic plasticity [7, 8] is becoming increasingly associated with malignant phenotypes [9]. Namely, mitochondrial oxidation (e.g. glucose metabolism through the tricarboxylic acid cycle, TCA) is linked with microenvironment adaptation and tumor progression [10].
The ability to use both glycolysis and mitochondrial oxidation pathways is a critical feature of GBM, which has been demonstrated from preclinical models to patients [11–13]. More recently, specific dependencies/proclivities towards those metabolic pathways are beginning to reveal GBM subtypes with prognostic value in human cell lines and patient-derived cells [14–16]. Importantly, the latest WHO classification of central nervous system tumors now distinguishes two metabolic phenotypes of adult GBM based on molecular assessment of a specific TCA cycle mutation (isocitrate dehydrogenase, IDH), namely into grade 2-4 gliomas (IDH-mut) and grade 4 GBM (IDH-wt) [17]. The prognostic value of GBM metabolic phenotypes clearly calls for non-invasive imaging methodologies capable of resolving the different subtypes, both for diagnosis and for treatment response monitoring. However, such methods are scarce.
Deuterium metabolic imaging (DMI) has been proposed for mapping active metabolism de novo in several tumor models [18–24]. While this has also been demonstrated in GBM patients [18], and more recently in mouse models of patient-derived GBM subtypes [25], mapping glucose metabolic fluxes remains unaddressed in these tumors due to the poor temporal resolution of DMI; particularly for glucose mitochondrial oxidation. Leveraging the benefits and risks of denoising methods for MR spectroscopy [26–28], we recently combined Deuterium Magnetic Resonance Spectroscopy (2H-MRS) [29] with Marcheku-Pastur PCA (MPPCA) denoising [30] to propose Dynamic Glucose-Enhanced (DGE) 2H-MRS [31], demonstrating its ability to quantify glucose fluxes through glycolysis and mitochondrial oxidation pathways in vivo in mouse GBM, which in turn revealed their proliferation status.
Here, we develop and apply a novel rapid DGE-DMI method to spatially resolve glucose metabolic flux rates in mouse GBM and reach a temporal resolution compatible with its kinetic modeling. For this, we adapt two advances of PCA denoising – tensor MPPCA [32, 33] and threshold PCA denoising [34] – and apply it for regional metabolic assessment of mouse GBM. First, we validated our novel approach in vivo for its ability to map glucose fluxes through glycolysis and mitochondrial oxidation in mouse GBM. Then, we investigate the potential of our new approach for depicting histopathologic differences in two mouse models of glioblastoma, including cell proliferation, peritumoral infiltration and migration. For this we used the same allograft mouse models of GBM, induced with CT2A and GL261 cell lines [35–39], but at more advanced stages of progression [31].
Results
MRI assessment of mouse GBM
Multi-parametric MRI provided a detailed characterization of each cohort at endpoint. Volumetric T2-weighted MRI indicated consistent tumor sizes across CT2A and GL261 cohorts (58.5±7.2 mm3). GL261 tumors were studied sooner after induction (17±0 vs 30±5 days post- injection, p=0.032), explaining the lower animal weights in this cohort (22.4±0.6 vs 25.7±0.9 g, p=0.017). DCE T1-weighted MRI indicated higher vascular permeability (0.85±0.11 vs 0.43±0.05 10-2/min, p=0.012) and a tendency for larger extracellular volume fractions (0.26±0.03 vs 0.18±0.02, p=0.056) in the GL261 tumors compared to CT2A. However, DCE T1-weighted MRI was carried out only in 80% of the mice due to time restrictions. This information is detailed in Table S1, where quantitative assessment of DGE-DMI, DCE-T1 and histologic parameters is displayed for tumor and tumor border regions, based on ROI analysis.
DGE-DMI in mouse GBM
Tumor metabolic assessment was performed with DGE-DMI in CT2A vs GL261 cohorts. No differences in RF coil quality or magnetic field homogeneity were detectable between the two cohorts: Q-factor 2H, 175±8 vs 176±9 (p=0.8996), respectively; FWHM 1H (VOI), 29.2±6.6 vs 26.0±4.3 Hz (p=0.3837), respectively. DGE-DMI was used to map the natural abundance semi- heavy water signal (DHO) as well as the dynamic conversion of deuterium-labelled glucose (Glc) to its downstream products, lactate (Lac) and glutamate-glutamine (Glx) pools, in tumor and peritumor brain regions (Fig. 1A). Tensor PCA denoising improved the spectral quality compared to the original data, without any depictable effects in the relative spatial distributions of SNR (Fig. S1), leading to a consistent and significant ∼3-fold SNR increase across all the subjects (from 6.4±0.1 before denoising to 20.1±0.4 after denoising, Table S1).
Spectral quantification of DGE-DMI data in each voxel and time point rendered time-course de novo concentration maps for each metabolite (DHO, Glc, Glx, and Lac), in both GBM cohorts (Fig 1B). Voxel-wise averaging of DGE-DMI time-course data after Glc injection generated average metabolic concentration maps for each tumor (Fig. 1C). Thus, Lac concentration was visually higher in the tumor regions, due to enhanced glycolysis; whereas Glx was more apparent in the adjacent non/peri-tumoral areas, consistent with a more prevalent oxidative metabolism in the normal brain. Kinetic fitting of DGE-DMI time-course concentration maps rendered glucose flux maps, namely its maximum consumption rate (Vmax) and flux rates through glycolysis (Vlac and klac) and mitochondrial oxidation (Vglx and kglx) (Fig 1D). Both cohorts displayed higher glycolytic metabolism in the tumors and more pronounced glucose oxidation in non-tumor regions, aligned with average concentration maps.
Histopathology assessment of GBM cohort differences
Histopathological analysis consisted of screening the CT2A and GL261 brain tumors for morphological features, including qualitative assessment of cell density, hemorrhage, tumor vessels, necrosis, quantification of peripheral infiltration and quantification of tumor proliferation index, while blinded to the in vivo MRI/MRS data – Table S2. Thus, tumors were scored individually for the following stromal-vascular phenotype, as in [31], where: pattern I corresponds to predominance of small vessels, complete endothelial cell lining and sparse hemorrhages; pattern II to vasodilation and marked multifocal hemorrhages; pattern III to predominance of necrosis of the vascular wall, incomplete endothelial cell lining, vascular leakage, and edematous stroma; and pattern IV to tumors with absence of clear vascular spaces and edematous stroma.
Stromal-vascular phenotypes reflected the more advanced stages of tumor progression in which these tumors were collected, as compared to our previous study [31]. Particularly, CT2A (n=5) presented patterns I to III, whereas all GL261 (n=5) matched pattern IV. Moreover, the increased infiltrative and migratory characteristics of GL261 compared to CT2A tumors were evident in their irregular tumor borders and higher incidence of secondary brain lesions (Fig 2A). These findings collectively suggest a more invasive and aggressive pattern of GL261 tumors, characterized by reduced cell-cell adhesion and enhanced migratory potential compared to CT2A. This difference between the two groups was further highlighted in the quantitative regional analysis, in which Tumor-to-Border ROI ratios exhibited 47% lower cell density (p=0.004) and 32% higher cell proliferation (p=0.026) in GL261 compared to CT2A (Fig 2B).
Despite the more advanced stages of tumor progression, the results were largely consistent with the marked morphological differences between the two models [31]: CT2A with dense, cohesive and homogeneous cell populations (Fig 2A, left-side); GL261 displaying marked heterogeneity, with poorly cohesive areas and more infiltrative growth (Fig 2A, right-side). Quantitative assessment (nuclear counts) further confirmed a nearly 2-fold lower cell density of GL261 tumors compared to CT2A (4.9 vs 8.2 ·103 cells/µm2, p<0.001) despite their similar proliferation index (Table S1); and tumor cell density correlated with cell proliferation, strongly for CT2A (R=0.96, p=0.009) and the same tendency detected for GL261 (R=0.74, p=0.151).
Tumor volume and whole-brain gross assessment of cell density, cell proliferation, and glucose metabolism also revealed strong inter-subject correlations in both cohorts (Fig. S2): de novo glutamate-glutamine accumulation decreased with tumor size (R CT2A/ GL261/ pooled: -0.597/ - 0.753/ -0.455), consistent with its role as marker of oxidative metabolism in the normal brain; lactate synthesis rate increased with cellularity (R CT2A/ GL261/ pooled: +0.921/ +0.685/ +0.852), also aligned with enhanced glycolysis in growing tumors; whereas glucose accumulation reflected cell proliferation (R CT2A/ GL261/ pooled: +0.469/ +0.528/ +0.440).
Regional assessment of glucose metabolism in the tumor microenvironment
Initial intra-tumor analysis of DGE-DMI and DCE-T1 maps (pixel-wise correlations in tumor ROIs) indicate stronger correlations between de novo lactate accumulation (Lac) and vascular permeability (ktrans) in both cohorts (R between [+0.306 +0.741]), and extracellular space (ve) to some extent (R between [-0.084 +0.804]) – both less apparent without tensor PCA denoising (R between [+0.089 +0.647] and [-0.160 +0.684], respectively) (Fig. S3). Such accumulation of lactate according to local vascular permeability mostly reflected regional differences in glycolytic fluxes (Vlac: R between [-0.066 +0.510]), rather than lactate elimination rates (klac: R between [- 0.643 +0.460]). No additional correlations were detected.
GL261 tumors accumulated significantly less lactate in the core (1.60±0.25 vs 2.91±0.33 mM: -45%, p=0.013) and border regions (0.94±0.09 vs 1.46±0.17 mM: -36%, p=0.025) than CT2A – Fig 3 A-B, Table S1. Lower tumor lactate levels were associated with higher lactate elimination rate, klac (0.11±0.1 vs 0.06±0.01 mM/min: +94%, p=0.006) – Fig 3B. Further analysis of tumor/border metabolic ratios (Table S3) revealed +38% glucose (p=0.002) and -17% lactate (p=0.038) concentrations, and +55% higher lactate consumption rate (p=0.040) in the GL261 cohort. Moreover, lactate elimination rate correlated inversely with “tumor age” (time post- induction) in pooled cohorts (R=-0.66, p=0.039), and more consistently with tumor vascular permeability (ktrans: R=0.78, p=0.022) (Fig 3 C), rather than washout rate (kep: R=0.61, p=0.109).
Association between glucose metabolism and peritumoral invasion and migration
Finally, we investigated the association between glucose metabolism and phenotypic features of tumor aggressiveness, namely cell proliferation and tumor cell invasion and migration associated with secondary brain lesions. Only the more infiltrative GL261 cohort displayed inter-subject associations between tumor cell proliferation (Ki67+ %) and metabolism, namely inverse correlations with tumor border/peritumoral glucose oxidation rate (Vglx: R=-0.91, p=0.030) and glucose-derived glutamate-glutamine elimination rate (kglx: R=-0.99, p<0.001). Regrouping subjects according to glioma cell invasion and migration concomitant with secondary brain lesions (presence: C1, G3, G4, G5; vs. absence: C2, C3, C4, C5, G1, G2) revealed lower de novo glutamate-glutamine levels in peritumor brain regions (Glx: -37%, p=0.013), which were associated with its higher elimination rate (kglx: +69%, p=0.012) – Fig 4.
Discussion
Glioblastomas are aggressive brain tumors with a poor prognosis, largely due to their inter- and intra-tumor heterogeneity and lack of non-invasive methods to assess it. Here we developed and applied a DGE-DMI approach capable of generating metabolic concentration maps and flux rates in two mouse models of glioblastoma, based on unambiguous spectral quantification according to quality criteria. Our results suggest that glycolytic lactate turnover mirrors phenotype differences between the two glioblastoma models, whereas glucose-derived glutamate-glutamine recycling could underly glioma cell migration leading to secondary lesions. This information became more readily available when using the tensor PCA method for spectral denoising.
Tensor PCA denoising increased spectral SNR by ∼3-fold, consistently improving spectral quality observed in tumor and peritumoral regions without altering the spatiotemporal profiles of the metabolic concentration maps (Fig S4). While this had no apparent effect on metabolic concentration maps (Figs S5-6), it significantly improved the kinetic modeling performance (Fig S7) and rendered better quality metabolic flux maps in CT2A and GL261 cohorts. Thus, 63% increased pixel detectability enabled capturing more spatial features in the latter without affecting parameter estimates or introducing group differences (Figs S8-9).
Gross whole brain analysis revealed strong inter-subject correlations in both cohorts, such as higher lactate synthesis rate with increasing cellularity – consistent with enhanced glycolysis in growing tumors – whereas intra-tumor pixel-wise analysis suggested lactate accumulation according to local vascular permeability, mostly associated with regional differences in glycolytic fluxes. Such pixel-wise analyses might be misleading since de novo lactate diffuses quickly within tumor extracellular spaces and peritumoral regions [40], with spatiotemporal dynamics not fully captured by DGE-DMI. Namely, water diffusion in GL261 tumors in vivo (apparent diffusion coefficient ∼10-3 mm2/s [41, 42]) extends beyond the in-plane voxel area (0.56×0.56 = 0.31 mm2) during each time frame (12 min). Thus, we focused instead on inter-tumor ROI analysis of glucose metabolic fluxes, in tumor and peritumoral (border) regions.
Compared to our previous study using the same GBM models [31], larger tumors (59±7 vs 38±3 mm3) display more disrupted stromal-vascular phenotypes (H&E scores: CT2A I-III vs I; GL261, IV vs I-IV) and weaker cell-cell interactions (lower cohesiveness) (Table S2), associated with lower vascular permeability (ktrans: 6±1 vs 14±1 103/min) and leading to lower glucose oxidation rates (Vglx: 0.28±0.06 vs. 0.40±0.08 mM/min), but remarkably similar glycolytic fluxes (Vlac: 0.59±0.04 vs. 0.55±0.07 mM/min). Thus, glycolysis flux rates are relatively well preserved across GL261 and CT2A mouse GBM models, regardless of tumor volume and vascular permeability.
GL261 tumors were examined earlier after induction than CT2A (17±0 vs. 30±5 days, p = 0.032), displaying similar volumes (57±6 vs. 60±14, p = 0.813) but better vascular permeability (8.5±1.1 vs 4.3±0.5 103/min: +98%, p=0.001), lower cell density (4.9±0.2 vs. 8.2±0.3 10-3 cells/µm2: -40%, p<0.001), and more disrupted stromal-vascular phenotypes and infiltrative growth (5/5 vs 0/5). Such GBM cohort differences were markedly reflected in their regional lactate metabolism. Thus, GL261 tumors accumulated roughly -40% less lactate in tumor and tumor border regions, associated with +94% higher lactate elimination rate rather than glycolytic rate differences in tumor regions, as could be assumed solely based on metabolic concentration maps.
Tumor core vs tumor border analyses further suggest that lactate metabolism reflects regional histologic differences between the two cohorts: lactate accumulation mirrors cell density gradients, whereas lactate consumption/elimination rate coarsely reflects differences in cell proliferation. This is consistent with GL261’s lower cell density and cohesiveness, more disrupted stromal-vascular phenotypes, and infiltrative growth pattern at the tumor border area, where relatively lower cell division is expected [43]. Altogether, our results suggest increased lactate consumption rate (active recycling) in GL261 tumors with higher vascular permeability, e.g. as a metabolic substrate for oxidative metabolism [44] promoting GBM cell survival and invasion [45]. While, lactate shuttling within the tumor microenvironment is also reported in other tumor types, between cancer cells [46] and between cancer and stromal cells [47, 48], it should be noted that oxidative phosphorylation inefficiency has been extensively documented in cancer cells, including GBM [49], largely associated with hypoxic niches and in agreement with our measurements of lower glucose oxidation rate (Vglx) in tumor vs. peritumoral regions.
The lower glucose oxidation rates measured in this study compared with smaller, better perfused tumor [31], are in good agreement with our previous data indicating quick adaptation of this pathway flux according to oxygen availability in the tumor microenvironment [31]. Under such physiological conditions – underlying more advanced progression stages, reflected in more disrupted stromal-vascular phenotypes – tumor glucose oxidation rate was not associated with cell proliferation index, consistent with previous observations [31]. Instead, tumor cell proliferation was inversely correlated with tumor border/peritumoral glucose oxidation rate and glucose-derived glutamate-glutamine elimination rate in more infiltrative GL261 tumors; but not in CT2A. This observation is consistent to some extent with GL261 cells’ and tumor’s ability to modulate mitochondrial metabolism according to their microenvironment (e.g. oxygen availability [31]), which is likely to occur during their progression from more circumscribed/local cell proliferation towards more disrupted stromal-vascular phenotypes with higher peritumoral infiltration and distant migration.
Notably, glucose-derived glutamate-glutamine displayed -37% lower levels and +69% higher elimination rate in peritumor regions of mouse brains bearing secondary GBM lesions (respective primary tumors displaying +146% increased glucose oxidation rate, detectable only with tensor PCA denoising – Fig S10). This could be associated with glutamate-glutamine-driven mitochondrial metabolism, through the TCA cycle coupled with oxidative phosphorylation (more prevalent in the normal brain) and/or via substrate level phosphorylation for ATP synthesis – glutaminolysis (as reported in glioma cells, e.g. CT2A [50]). While patient-derived xenograft models would be more suited to recapitulate human GBM infiltration, our observations are well aligned with the pivotal role of mitochondrial metabolism in cancer cells with higher motile potential, as reported in human GBM [51] and in mouse and human breast cancer cell lines [52, 53]. Particularly, the dynamics of glutamate shuttling underlying neuronal-glioma cell communication and promoting GBM infiltration, are increasingly reported by the emerging field of cancer neuroscience [54]. Therefore, our results suggest that glucose mitochondrial metabolism mirrors GBM progression in mouse GL261 and CT2A models: more prevalent in smaller, well perfused tumors, where glucose oxidation rate correlates with tumor cell proliferation [31]; lower in larger, more poorly perfused tumors, where glutamate-glutamine recycling may reflect a phenotype associated with secondary brain lesions.
Despite the excellent performance of tensor PCA denoising – 3-fold increase in SNR, approaching the original/raw values obtained previously with single-voxel 2H-MRS data (SNR∼20, [31]) – no further improvements in SNR could be achieved by FID averaging within the tumor ROI (Fig S11). Therefore, further DGE-DMI preclinical studies aimed at detecting and quantifying relatively weak signals, such as tumor glutamate-glutamine, should improve basal SNR with higher magnetic field strengths, more sensitive RF coils, and advanced DMI pulse sequences [55]). In the kinetic model, the extracellular volume fraction was fixed to ensure model stability, as previously demonstrated using the tumor average across all subjects [31]. This approximation may not fully reflect the intra- and inter-tumor heterogeneity of this parameter in both cohorts, and may not be representative of its peritumoral regions. Still, we opted for this approach, rather than pixel-wise adjustments according to DGE-T1 extracellular volume fraction maps, given (i) the relative insensitivity of the model to the actual extracellular volume fraction value used [31], also verified in the present study (Fig S12); and particularly, because (ii) we did not have DCE-T1 data for the full cohort, thus it was not feasible to perform individual corrections, which in any case would ultimately be prone to error at tumor periphery/border regions, where exact delimitations are typically debatable.
In summary, DGE-DMI quantitatively maps glycolysis and mitochondrial oxidation fluxes in mouse GBM, highlighting its importance for metabolic characterization and potential for in vivo GBM phenotyping. In large tumors, lactate metabolism underlies mouse GBM model-specific features, consistent with faster turnover in more disrupted stromal-vascular phenotypes and mirroring intra-tumor gradients of cell density and proliferation, whereas glutamate-glutamine recycling may reflect a phenotype associated with secondary brain lesions. Tensor PCA denoising significantly improved spectral signal-to-noise, which helped reveal such associations between regional glucose metabolism and phenotypic features of intra- and inter-tumor heterogeneity. These results clearly highlight the importance of mapping pathway fluxes alongside de novo concentrations to improve the characterization of the complex and dynamic heterogeneity of GBM metabolism. This may be determinant for selecting among new treatment modalities targeting GBM metabolism [56, 57] or monitoring the efficacy of novel immunotherapy approaches [58] beyond conventional chemoradiotherapy [25]. Importantly, DGE-DMI is potentially translatable to high-field clinical MRI scanners, as already demonstrated for DMI at 9.4T [59], benefiting from the higher sensitivity in the much larger human brain compared to mice: 200 cm3 [60] and 415 mm3 [61], respectively.
Materials and Methods
Animals and cell lines
All animal experiments were pre-approved by the competent institutional as well as national authorities, and carried out strictly adhering to European Directive 2010/63. A total of n=10 C57BL/6j male mice were used in this study, bred at the Champalimaud Foundation Vivarium, and housed with ad libitum access to food and water and 12h light cycles. GL261 mouse glioma cells were obtained from the Tumor Bank Repository at the National Cancer Institute (Frederick MD, USA). CT2A mouse glioma cells were kindly provided by Prof. Thomas Seyfried at Boston College (Boston MA, USA). Both cell lines were grown in RPMI-1640 culture medium supplemented with 2.0 g/l Sodium Bicarbonate, 0.285 g/l L-glutamine, 10% Fetal Bovine Serum (Gibco) and 1% Penicillin-Streptomycin solution. The cell lines tested negative for mycoplasma contamination using the IMPACT Mouse FELASA 1 test (Idexx-BioResearch, Ludwigsburg, Germany).
Glioma models
Tumors were induced in previously described [62]. Briefly, intracranial stereotactic injection of 1 x105 GL261 or CT2A cells was performed in the caudate nucleus (n=5 and n=5 mice, respectively); analgesia (Meloxicam 1.0 mg/Kg s.c.) was administered 30 min before the procedure. Mice were anesthetized with isoflurane (1.5-2.0% in air) and immobilized on a stereotactic holder (Kopf Instruments, Tujunga/CA, USA) where they were warmed on a heating pad at 37 °C, while body temperature was monitored with a rectal probe (WPI ATC-2000, Hitchin, UK). The head was shaved with a small trimmer, cleaned with iodopovidone, and the skull exposed through an anterior-posterior incision in the midline with a scalpel. A 1 mm hole was drilled in the skull using a micro-driller, 0.1 mm posterior to the bregma and 2.32 mm lateral to the midline. The tumor cells (1x105 in 4 μL PBS) were inoculated 2.35 mm below the cortical surface using a 10 µL Hamilton syringe (Hamilton, Reno NV, USA) connected to an automatic push-pull microinjector (WPI SmartouchTM, Sarasota FL, USA), by advancing the 26G needle 3.85 mm from the surface of the skull (∼1mm skull-to-brain surface distance), pulling it back 0.5 mm, and injecting at 2 μL/min rate. The syringe was gently removed 2 min after the injection had finished, the skin sutured with surgical thread (5/0 braided silk, Ethicon, San Lorenzo Puerto Rico) and wiped with iodopovidone. During recovery from anesthesia, animals were kept warm on a heating pad and given an opioid analgesic (Buprenorphine 0.05 mg/Kg s.c.) before returning to their cage. Meloxicam analgesia was repeatedly administered at 24- and 48-hours post-surgery.
In vivo Studies
Longitudinal MRI
GBM-bearing mice were imaged every 5-7 days on a 1 Tesla Icon MRI scanner (Bruker BioSpin, Ettlingen, Germany; running ParaVision 6.0.1 software), to measure tumor volumes. For this, each mouse was placed in the animal holder under anesthesia (1-2 % isoflurane in 31% O2), heated with a recirculating water blanket, and monitored for rectal temperature (36-37 °C) and breathing (60-90 BPM). Tumor volume was measured with T2-weighted 1H-MRI (RARE sequence, ×8 acceleration factor, repetition time TR = 2500 ms, echo time TE = 84 ms, 8 averages, 1 mm slice thickness, and 160×160 µm2 in-plane resolution), acquired in two orientations (coronal and axial). Each session lasted up to 30 min/animal.
End-point MRI and DMI
GBM-bearing mice with tumors ≥35 mm3 (longitudinal MRI assessment) were scanned on a 9.4T BioSpec MRI scanner (Bruker BioSpin, Ettlingen, Germany; running under ParaVision 6.0.1), using a 2H/1H transmit-receive surface coilset customized for the mouse brain (NeosBiotec, Pamplona, Spain), as described before [31]. Before each experiment, GBM-bearing mice fasted 4- 6h, were weighed, and cannulated in the tail vein with a catheter connected to a home-built 3-way injection system filled with: 6,6′-2H2-glucose (1.6M in saline); Gd-DOTA (25 mM in saline); and with heparinized saline (10 U/mL). Mice were placed on the animal holder under anesthesia (as in 2.3.1). Coilset quality factors (Q) for 1H and 2H channels were estimated in the scanner for each sample based on the ratio of the resonance frequency (400.34 and 61.45 MHz, for protons and deuterium, respectively) to its bandwidth (full width at half-minimum of the wobbling curve during the initial tuning adjustments): 175±8 and 200±12, respectively. Mice were imaged first with T2-weighted 1H-MRI (RARE sequence, x8 acceleration factor, 3000 ms TR, 40 ms TE; 2 averages, 1 mm slice thickness, 70 µm in-plane resolution) in two orientations (coronal and axial). Then, the magnetic field homogeneity was optimized over the tumor region based on the water peak with 1H-MRS (STEAM localization: 6x6x3 mm volume of interest, i.e. 108 µL) using localized 1st and 2nd order shimming with the MapShim Bruker macro, leading to full widths at half-maximum (FWHM) of 28±5 Hz.
DMI was performed using a slice-FID chemical-shift imaging pulse sequence, with 175 ms TR, 256 spectral points sampled over a 1749 Hz window, and Shinnar-Le Roux RF pulse [63, 64] (0.42ms, 10kHz) with 55° flip angle, to excite a brain slice including the tumor: 18×18 mm field- of-view, and 2.27 mm slice thickness. After RF pulse calibration (using the natural abundance semi-heavy water peak, DHO), DGE-DMI data were acquired for 2h23min (768 repetitions), with i.v. bolus of 6,6′-2H2-glucose (2 g/Kg, injected over 30 s; Euroisotop, St Aubin Cedex, France). Data were sampled with an 8×8 matrix and 4-fold Fourier interpolated, rendering a 560 µm in-plane resolution. A reference T2-weighted image was additionally acquired with matching field- of-view and slice thickness, and 70 µm in-plane resolution.
Finally, animals underwent DCE T1-weighted 1H-MRI (FLASH sequence, 8° flip-angle, 16ms TR, 4 averages, 150 repetitions, 1 slice with 140 µm in-plane resolution and 2.27 mm thickness, FOV size and position matching the DGE-DMI experiment), with i.v. bolus injection of Gd-DOTA (0.1 mmol/Kg, injected over 30 s; Guerbet, Villepinte, France). Animals were then sacrificed, brains were removed, washed in PBS, and immersed in 4% PFA.
MRI/DMI Processing
T2-weighted 1H-MRI
T2-weighted MRI data were processed in ImageJ 1.53a (Rasband, W.S., ImageJ, U. S. National Institutes of Health, Bethesda, Maryland, USA, https://imagej.nih.gov/ij/, 1997-2018). For each animal, the tumor region was manually delineated on each slice, and the sum of the areas multiplied by the slice thickness to estimate the volume, which was averaged across the two orientations acquired (coronal and axial).
DGE-DMI
DGE-DMI data were processed in MATLAB® R2018b (Natick, Massachusetts: The MathWorks Inc.) and jMRUI 6.0b [65]. Each dataset was averaged to 12 min temporal resolution and the central spectral matrix region selected (to discard noise regions outside the brain, as well as the olfactory bulb and cerebellum), rendering a 4D spectral-spatial-temporal matrix of 256×32×32×12 points. After automated phase-correction of each spectrum, the 4D matrix was denoised with a tensor PCA denoising approach [32]. For this, a [8 8 8] window and tensor structure [1 2:3 4] were used for patch processing the spectral, spatial, and temporal dimensions with, whereas the a priori average standard deviation of the noise in each spectrum (calculated σ2) was used to avoid deleterious effects of spatially-correlated noise [34]. Then, these denoised spectra were analyzed voxel-wise by individual peak fitting with AMARES (similarly to the single-spectrum analysis reported previously in [52]), using a basis set for DHO (4.76 ppm: short- and long-T2 fractions [18]) and deuterium-labelled: glucose (Glc, 3.81 ppm), glutamate-glutamine (Glx, 2.36 ppm), and lactate (Lac, 1.31 ppm); relative linewidths referenced to the estimated short- T2 fraction of DHO, according to the respective T2 relaxation times reported by de Feyter et al [18]. The natural abundance DHO peak (DHOi) was further used to select and quantify both original and denoised spectra: SNRDHOi >3.5 and 13.88 mM reference (assuming 80 % water content in the brain and 0.03 % natural abundance of DHO), respectively. Metabolite concentrations (CRLB<50%; otherwise discarded) were corrected for T1 and labeling-loss effects, according to the values reported by de Feyter et al (T1, ms: DHO, 320; Glc, 64; Glx, 146; Lac, 297) [18] and de Graaf et al (number of magnetically equivalent deuterons: DHO, 1; Glc, 2; Glx, 1.2; Lac, 1.7) [66], respectively. Thus, the concentration of each metabolite (m) at each time point was estimated as (Eq 1):
Area = peak area; Area0 = average peak area before injection; d = number of magnetically equivalent deuterons corrected for labelling-loss effects; C = T1 correction factor (1-exp(-TR/T1)); and Concref = reference DHO concentration.
The time-course changes of 2H-labelled metabolite (Glc, Glx and Lac) concentrations were fitted using a modified version of the kinetic model reported by Kreis et al [19], to estimate the maximum rate of Glc consumption (total, Vmax) for Glx synthesis (mitochondrial oxidation, Vglx) and Lac synthesis (glycolysis, Vlac), and the confidence intervals for all estimated parameters:
The coupled differential equations describing the concentration kinetics of each metabolite were:
where: kg, apparent rate constant of glucose transfer between blood and tumor (min−1); kglx, apparent rate constant of Glx elimination (min−1); klac, apparent rate constant of lactate elimination (min−1); Cp = a1 · e−kp·t, Glc concentration in plasma (mM); a1, the Glc concentration after the bolus injection (mM); and kp, the effective rate constant of labeled glucose transfer to tissue (min−1). As reported previously [31], the following parameters were fixed: fraction of deuterium enrichment (f), at 0.6 [19]; constant for glucose uptake (km), at 10 mM [67, 68]; and the extravascular-extracellular volume fraction (v), at 0.22 – average estimation from DCE-T1- weighted MRI analysis (Table S1). All the other parameters were fitted without any restrictions to their range.
DCE T1-weighted MRI
DCE T1-weighted MRI data were processed with DCE@urLab [69], as before [31]. First, ROIs were manually delineated for each tumor and the time-course data was fitted with the Extended Tofts 2-compartment model [70], to derive the volume transfer constant between plasma and tumor extravascular-extracellular space (ktrans), the washout rate between extravascular-extracellular space and plasma (kep), and the extravascular-extracellular volume fraction (ve). Then, each dataset was reprocessed by down-sampling the original in-plane resolution to match the DGE-DMI experiment (0.56×0.56×2.27 mm3), and fitting the time-course data pixel-wise with the Extended Tofts 2-compartment model to derive ktrans, kep, and ve maps (pixels with root-mean square error >0.005 discarded).
Histopathology and Immunohistochemistry
Whole brains fixed in 4% PFA were embedded in paraffin and sectioned at 30 different levels on the horizontal plane, spanning the whole tumor area. 4 µm sections were stained with H&E (Sigma-Aldrich, St. Louis MO, USA), digitized (Nanozoomer, Hamamatsu, Japan), and analyzed by an experimental pathologist blinded to experimental groups, according to previously established criteria [31]. Then, QuPath v0.4.3 built-in tools [71] were used to highlight different tumor regions: Tumor ROIs, corresponding to the bulk tumor, were delineated first with “create threshold” and then manually corrected; Border ROIs, including areas of peritumoral infiltration, were delineated with “expand annotations” by expanding 100 µm the tumor margin toward the adjacent brain parenchyma; Infiltrative ROIs, corresponding to specific infiltrative regions, were manually annotated. Between 3 to 6 sections of each tumor were also immunostained for Ki67 (mouse anti-ki67, BD, San Jose CA, USA; blocking reagent, M.O.M ImmPRESS kit, Vector Laboratories, Burlingame CA, USA; liquid DAB+, Dako North America Inc, Carpinteria CA, USA), digitized (Nanozoomer, Hamamatsu, Japan), and analyzed with QuPath built-in tools [71] for Tumor and Border ROIs, defined as detailed above. Thus, Ki67+/- cells were counted semi- automatically to determine the total number of cells, the cell density, and the proliferation index (% Ki67+ cells) as the average across slices for each ROI, and respective tumor/border ratios. This procedure was repeated for each animal.
Statistical analyses
Data were analyzed in MATLAB® R2018b (Natick, Massachusetts: The MathWorks Inc.) using the two-tailed Student’s t-test, either unpaired (comparing different animal cohorts) or paired (comparing the same animal cohort in different conditions). Differences at the 95% confidence level (p=0.05) were considered statistically significant. Correlation analyses were carried out with the Pearson R coefficient. Error bars indicate standard deviation unless indicated otherwise.
Acknowledgements
This work was supported by: H2020-MSCA-IF-2018, ref. 844776 (RVS); FCT CEEC- IND4ed, ref 2021.02777.CEECIND/CP1675/CT0003 (RNH); and the Champalimaud Foundation. The authors thank Dr. Thomas Seyfried for access to the CT2A cell line and helpful discussion, and the Vivarium of the Champalimaud Centre for the Unknown, a research infrastructure of CONGENTO co-financed by Lisbon Regional Operational Programme (Lisboa2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF) and Fundação para a Ciência e Tecnologia (Portugal), under the project LISBOA-01- 0145-FEDER-022170.
References
- 1.Malignant gliomas in adultsN Engl J Med 359:492–507
- 2.Evolutionary dynamics of carcinogenesis and why targeted therapy does not workNat Rev Cancer 12:487–93
- 3.The metabolic cooperation between cells in solid cancer tumorsBiochim Biophys Acta 1846:216–25
- 4.The Emerging Hallmarks of Cancer MetabolismCell Metab 23:27–47
- 5.On the origin of cancer cellsScience 123:309–14
- 6.Why do cancers have high aerobic glycolysis?Nat Rev Cancer 4:891–9
- 7.Cancer’s Fuel Choice: New Flavors for a Picky EaterMol Cell 60:514–23
- 8.Metabolomic changes accompanying transformation and acquisition of metastatic potential in a syngeneic mouse mammary tumor modelJ Biol Chem 285:9317–21
- 9.Metabolic Plasticity as a Determinant of Tumor Growth and MetastasisCancer Res 76:5201–8
- 10.Metabolic reprogramming and cancer progressionScience 368
- 11.Acetate is a bioenergetic substrate for human glioblastoma and brain metastasesCell 159:1603–14
- 12.Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastomaNat Cell Biol 17:1556–68
- 13.Metabolism of [U-13 C]glucose in human brain tumors in vivoNMR Biomed 25:1234–44
- 14.Integrated genetic and metabolic landscapes predict vulnerabilities of temozolomide resistant glioblastoma cellsNPJ Syst Biol Appl 7
- 15.Pathway-based classification of glioblastoma uncovers a mitochondrial subtype with therapeutic vulnerabilitiesNature Cancer 2:141–56
- 16.Beyond the Warburg Effect: Oxidative and Glycolytic Phenotypes Coexist within the Metabolic Heterogeneity of GlioblastomaCells 10
- 17.The 2021 WHO Classification for Gliomas and Implications on Imaging Diagnosis: Part 1-Key Points of the Fifth Edition and Summary of Imaging Findings on Adult-Type Diffuse GliomasJ Magn Reson Imaging 58:677–689
- 18.Deuterium metabolic imaging (DMI) for MRI-based 3D mapping of metabolism in vivoSci Adv 4
- 19.Measuring Tumor Glycolytic Flux in Vivo by Using Fast Deuterium MRIRadiology 294:289–296
- 20.Monitoring tumor cell death in murine tumor models using deuterium magnetic resonance spectroscopy and spectroscopic imagingProc Natl Acad Sci U S A 118
- 21.Mapping of exogenous choline uptake and metabolism in rat glioblastoma using deuterium metabolic imaging (DMI)Front Cell Neurosci 17
- 22.Parallel detection of multi-contrast MRI and Deuterium Metabolic Imaging (DMI) for time-efficient characterization of neurological diseasesmedRxiv
- 23.Deuterium metabolic imaging reports on TERT expression and early response to therapy in cancerClin Cancer Res
- 24.High-sensitivity deuterium metabolic MRI differentiates acute pancreatitis from pancreatic cancers in murine modelsSci Rep 13
- 25.Deuterium metabolic imaging differentiates glioblastoma metabolic subtypes and detects early response to chemoradiotherapyCancer Res
- 26.Effects of apodization smoothing and denoising on spectral fittingMagn Reson Imaging 70:108–114
- 27.Uncertainty in denoising of MRSI using low-rank methodsMagn Reson Med 87:574–588
- 28.Denoising single MR spectra by deep learning: Miracle or mirage?Magn Reson Med 90:1749–1761
- 29.Quantitative assessment of brain glucose metabolic rates using in vivo deuterium magnetic resonance spectroscopyJ Cereb Blood Flow Metab 37:3518–3530
- 30.Denoising of diffusion MRI using random matrix theoryNeuroimage 142:394–406
- 31.Glucose fluxes in glycolytic and oxidative pathways detected in vivo by deuterium magnetic resonance spectroscopy reflect proliferation in mouse glioblastomaNeuroimage Clin 33
- 32.Tensor denoising of multidimensional MRI dataMagn Reson Med 89:1160–1172
- 33.A user independent denoising method for x-nuclei MRI and MRSMagn Reson Med 90:2539–2556
- 34.Efficient PCA denoising of spatially correlated redundant MRI dataImaging Neuroscience 1:1–26
- 35.Vascular apoptosis and involution in gliomas precede neovascularization: a novel concept for glioma growth and angiogenesisLab Invest 80:837–49
- 36.Studies in carcinogenesis. VIII. Experimental production of brain tumors in mice with methylcholanthreneAm J Cancer 37:364–395
- 37.Immunocompetent murine models for the study of glioblastoma immunotherapyJ Transl Med 12
- 38.Ganglioside distribution in murine neural tumorsMol Chem Neuropathol 17:147–67
- 39.Standardization of an orthotopic mouse brain tumor model following transplantation of CT-2A astrocytoma cellsHistol Histopathol 22:1309–26
- 40.Serial in vivo spectroscopic nuclear magnetic resonance imaging of lactate and extracellular pH in rat gliomas shows redistribution of protons away from sites of glycolysisCancer Res 67:7638–45
- 41.Preliminary characterization of an experimental breast cancer cells brain metastasis mouse model by MRI/MRSMagn Reson Mater Phy (MAGMA 21:237–49
- 42.Noninvasive diffusion magnetic resonance imaging of brain tumour cell size for the early detection of therapeutic responseSci Rep 10
- 43.Single-Cell RNA-Seq Analysis of Infiltrating Neoplastic Cells at the Migrating Front of Human GlioblastomaCell Rep 21:1399–1410
- 44.Lactate is an epigenetic metabolite that drives survival in model systems of glioblastomaMol Cell 82:3061–3076
- 45.Metabolic targeting of lactate efflux by malignant glioma inhibits invasiveness and induces necrosis: an in vivo studyNeoplasia 13:620–32
- 46.Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in miceJ Clin Invest 118:3930–42
- 47.Tumor stroma interaction is mediated by monocarboxylate metabolismExp Cell Res 352:20–33
- 48.Lactate influx through the endothelial cell monocarboxylate transporter MCT1 supports an NF-kappaB/IL-8 pathway that drives tumor angiogenesisCancer Res 71:2550–60
- 49.Could Cytoplasmic Lipid Droplets be Linked to Inefficient Oxidative Phosphorylation in Cancer?Current Tissue Microenvironment Reports
- 50.Mitochondrial Substrate-Level Phosphorylation as Energy Source for Glioblastoma: Review and HypothesisASN Neuro 10
- 51.Glioblastoma cell motility depends on enhanced oxidative stress coupled with mobilization of a sulfurtransferaseCell Death Dis 13
- 52.Metabolic plasticity of metastatic breast cancer cells: adaptation to changes in the microenvironmentNeoplasia 17:671–84
- 53.Mitochondrial Metabolism Drives Low-density Lipoprotein- induced Breast Cancer Cell MigrationCancer Res Commun 3:709–724
- 54.Glioblastoma hijacks neuronal mechanisms for brain invasionCell 185:2899–2917
- 55.Improving deuterium metabolic imaging (DMI) signal-to-noise ratio by spectroscopic multi-echo bSSFP: A pancreatic cancer investigationMagn Reson Med 86:2604–2617
- 56.An inhibitor of oxidative phosphorylation exploits cancer vulnerabilityNat Med 24:1036–1046
- 57.Gboxin is an oxidative phosphorylation inhibitor that targets glioblastomaNature 567:341–346
- 58.rWTC-MBTA Vaccine Induces Potent Adaptive Immune Responses Against Glioblastomas via Dynamic Activation of Dendritic CellsAdv Sci (Weinh 11
- 59.Deuterium metabolic imaging in the human brain at 9.4 Tesla with high spatial and temporal resolutionNeuroimage 244
- 60.Deciphering laminar-specific neural inputs with line-scanning fMRINat Methods 11:55–8
- 61.A three-dimensional MRI atlas of the mouse brain with estimates of the average and variabilityCereb Cortex 15:639–45
- 62.Perturbation of mouse glioma MRS pattern by induced acute hyperglycemiaNMR Biomed 21:251–64
- 63.The synthesis of soft pulses with a specified frequency responseMagn Reson Med 12:88–92
- 64.Parameter relations for the Shinnar-Le Roux selective excitation pulse design algorithm [NMR imaging]IEEE Trans Med Imaging 10:53–65
- 65.Quantitation of magnetic resonance spectroscopy signals: the jMRUI software packageMeasurement Science & Technology 20
- 66.Characterization of Kinetic Isotope Effects and Label Loss in Deuterium-Based Isotopic Labeling StudiesACS Chem Neurosci 12:234–243
- 67.Modeling cancer glycolysisBiochim Biophys Acta 1807:755–67
- 68.Quantitation of glucose uptake in tumors by dynamic FDG-PET has less glucose bias and lower variability when adjusted for partial saturation of glucose transportEJNMMI Res 2
- 69.DCE@urLAB: a dynamic contrast-enhanced MRI pharmacokinetic analysis tool for preclinical dataBMC Bioinformatics 14
- 70.Modeling tracer kinetics in dynamic Gd-DTPA MR imagingJ Magn Reson Imaging 7:91–101
- 71.QuPath: Open source software for digital pathology image analysisSci Rep 7
Article and author information
Author information
Version history
- Sent for peer review:
- Preprint posted:
- Reviewed Preprint version 1:
Copyright
© 2024, Simões et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.