1
|
Ren Y, Zhu L, Guo Y, Ma J, Yang L, Zheng C, Dong X. Melatonin enhances the efficacy of anti-PD-L1 by improving hypoxia in residual tumors after insufficient radiofrequency ablation. J Pharm Anal 2024; 14:100942. [PMID: 39263355 PMCID: PMC11388694 DOI: 10.1016/j.jpha.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 09/13/2024] Open
Abstract
The hypoxic microenvironment and inflammatory state of residual tumors caused by insufficient radiofrequency ablation (iRFA) are major reasons for rapid tumor progression and pose challenges for immunotherapy. We retrospectively analyzed the clinical data of patients with hepatocellular carcinoma (HCC) treated with RFA and observed that iRFA was associated with poor survival outcomes and progression-free survival. Using an orthotopic HCC mouse model and a colorectal liver metastasis model, we observed that treatment with melatonin after iRFA reduced tumor growth and metastasis and achieved the best outcomes when combined with anti-programmed death-ligand 1 (anti-PD-L1) therapy. In mechanism, melatonin inhibited the expression of epithelial-mesenchymal transitions, hypoxia-inducible factor (HIF)-1α, and PD-L1 in tumor cells after iRFA. Flow cytometry revealed that melatonin reduced the proportion of myeloid-derived suppressor cells and increased the proportion of CD8+ T cells. Transcriptomic analysis revealed an upregulation of immune-activated function-related genes in residual tumors. These findings demonstrated that melatonin can reverse hypoxia and iRFA-induced inflammation, thereby overcoming the immunosuppressive tumor microenvironment (TME) and enhancing the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Licheng Zhu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinqiang Ma
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangjun Dong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
2
|
Erbani J, Boon M, Akkari L. Therapy-induced shaping of the glioblastoma microenvironment: Macrophages at play. Semin Cancer Biol 2022; 86:41-56. [PMID: 35569742 DOI: 10.1016/j.semcancer.2022.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 01/27/2023]
Abstract
The intricate cross-talks between tumor cells and their microenvironment play a key role in cancer progression and resistance to treatment. In recent years, targeting pro-tumorigenic components of the tumor microenvironment (TME) has emerged as a tantalizing strategy to improve the efficacy of standard-of-care (SOC) treatments, particularly for hard-to-treat cancers such as glioblastoma. In this review, we explore how the distinct microenvironmental niches characteristic of the glioblastoma TME shape response to therapy. In particular, we delve into the interplay between tumor-associated macrophages (TAM) and glioblastoma cells within angiogenic and hypoxic niches, and interrogate their dynamic co-evolution upon SOC therapies that fuels malignancy. Resolving the complexity of therapy-induced alterations in the glioblastoma TME and their impact on disease relapse is a stepping stone to identify targetable pro-tumorigenic pathways and TAM subsets, and may open the way to efficient combination therapies that will improve clinical outcomes.
Collapse
Affiliation(s)
- Johanna Erbani
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Menno Boon
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Leila Akkari
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
3
|
She L, Su L, Liu C. Bevacizumab combined with re-irradiation in recurrent glioblastoma. Front Oncol 2022; 12:961014. [PMID: 36046037 PMCID: PMC9423039 DOI: 10.3389/fonc.2022.961014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
Background Glioblastoma is characterized by rich vasculature and abnormal vascular structure and function. Currently, there is no standard treatment for recurrent glioblastoma (rGBM). Bevacizumab (BEV) has established role of inhibiting neovascularization, alleviating hypoxia in the tumor area and activating the immune microenvironment. BEV may exert synergistic effects with re-irradiation (re-RT) to improve the tumor microenvironment for rGBM. Purpose The purpose of this study was to evaluate the safety, tolerability, and efficacy of a combination of BEV and re-RT for rGBM treatment. Methods In this retrospective study, a total of 26 rGBM patients with surgical pathologically confirmed glioblastoma and at least one event of recurrence were enrolled. All patients were treated with re-RT in combination with BEV. BEV was administered until progression or serious adverse events. Results Median follow-up was 21.9 months for all patients, whereas median progression-free survival (PFS) was 8.0 months (95% confidence interval [CI]: 6.5–9.5 months). In addition, the 6-month and 1-year PFS rates were 65.4% and 28.2%, respectively. The median overall survival (OS), 6-month OS rate, and 1-year OS rate were 13.6 months (95% CI: 10.2–17.0 months), 92.3%, and 67.5%, respectively. The patient showed good tolerance during the treatment with no grade > 3 grade side event and radiation necrosis occurrence rate of 0%. Combined treatment of gross total resection (GTR) before re-RT and concurrent temozolomide during re-RT was an independent prognostic factor that affected both OS and PFS in the whole cohort (OS: 0.067, 95% CI: 0.009–0.521, p = 0.010; PFS: 0.238, 95% CI: 0.076–0.744, p = 0.038). Conclusion In this study, re-RT combined with concurrent and maintenance BEV treatment was safe, tolerable, and effective in rGBM patients. Moreover, GTR before re-RT and selective concurrent temozolomide could further improve patient PFS and OS.
Collapse
Affiliation(s)
- Lei She
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Su
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
4
|
Simultaneous Recording of the Uptake and Conversion of Glucose and Choline in Tumors by Deuterium Metabolic Imaging. Cancers (Basel) 2021; 13:cancers13164034. [PMID: 34439188 PMCID: PMC8394025 DOI: 10.3390/cancers13164034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Tumors increase their glucose and choline uptake to support growth. These properties are employed to detect and identify tumors in the body by imaging the uptake of radio-isotope analogs of these compounds. In this study we show that deuterium metabolic imaging (DMI) (a new MRI method to image metabolites using non-radioactive labeling with deuterium) can image choline uptake in tumors. Furthermore, we demonstrate that DMI can image the tumor uptake of choline and glucose (and additionally its metabolic conversion) simultaneously, in contrast to radio-isotope imaging, which only assesses the uptake of one radio-isotope labeled compound at a time. For these reasons (and also because DMI is relatively simple and can be combined with other MR methods), it is a promising modality for a more specific tumor characterization than by separate imaging of the uptake of radio-isotope labeled glucose or choline. Abstract Increased glucose and choline uptake are hallmarks of cancer. We investigated whether the uptake and conversion of [2H9]choline alone and together with that of [6,6′-2H2]glucose can be assessed in tumors via deuterium metabolic imaging (DMI) after administering these compounds. Therefore, tumors with human renal carcinoma cells were grown subcutaneously in mice. Isoflurane anesthetized mice were IV infused in the MR magnet for ~20 s with ~0.2 mL solutions containing either [2H9]choline (0.05 g/kg) alone or together with [6,6′-2H2]glucose (1.3 g/kg). 2H MR was performed on a 11.7T MR system with a home-built 2H/1H coil using a 90° excitation pulse and 400 ms repetition time. 3D DMI was recorded at high resolution (2 × 2 × 2 mm) in 37 min or at low resolution (3.7 × 3.7 × 3.7 mm) in 2:24 min. Absolute tissue concentrations were calculated assuming natural deuterated water [HOD] = 13.7 mM. Within 5 min after [2H9]choline infusion, its signal appeared in tumor spectra representing a concentration increase to 0.3–1.2 mM, which then slowly decreased or remained constant over 100 min. In plasma, [2H9]choline disappeared within 15 min post-infusion, implying that its signal arises from tumor tissue and not from blood. After infusing a mixture of [2H9]choline and [6,6′-2H2]glucose, their signals were observed separately in tumor 2H spectra. Over time, the [2H9]choline signal broadened, possibly due to conversion to other choline compounds, [[6,6′-2H2]glucose] declined, [HOD] increased and a lactate signal appeared, reflecting glycolysis. Metabolic maps of 2H compounds, reconstructed from high resolution DMIs, showed their spatial tumor accumulation. As choline infusion and glucose DMI is feasible in patients, their simultaneous detection has clinical potential for tumor characterization.
Collapse
|
5
|
Intratumoral Distribution of Lactate and the Monocarboxylate Transporters 1 and 4 in Human Glioblastoma Multiforme and Their Relationships to Tumor Progression-Associated Markers. Int J Mol Sci 2020; 21:ijms21176254. [PMID: 32872409 PMCID: PMC7504270 DOI: 10.3390/ijms21176254] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Metabolic reprogramming has been postulated to be one of the hallmarks of cancer, thus representing a promising therapeutic target also in glioblastoma multiforme (GBM). Hypoxic tumor cells produce lactate, and monocarboxylate transporters (MCTs) play an important role in its distribution; (2) Methods: We examined the distribution of lactate by multi voxel magnetic resonance spectroscopic imaging and ELISA in glioblastoma multiforme (GBM) patients. In addition, we investigated the expression and cellular localization of MCT1, MCT4, and of several markers connected to tumor progression by quantitative PCR and immunofluorescence double-staining in human GBM ex vivo tissues; (3) Results: The highest lactate concentration was found at the center of the vital parts of the tumor. Three main GBM groups could be distinguished according to their regional gene expression differences of the investigated genes. MCT1 and MCT4 were found on cells undergoing epithelial to mesenchymal transition and on tumor stem-like cells. GBM cells revealing an expression of cellular dormancy markers, showed positive staining for MCT4; (4) Conclusion: Our findings indicate the existence of individual differences in the regional distribution of MCT1 and MCT4 and suggest that both transporters have distinct connections to GBM progression processes, which could contribute to the drug resistance of MCT-inhibitors.
Collapse
|
6
|
Predicting Glioblastoma Response to Bevacizumab Through MRI Biomarkers of the Tumor Microenvironment. Mol Imaging Biol 2020; 21:747-757. [PMID: 30361791 DOI: 10.1007/s11307-018-1289-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE Glioblastoma (GB) is one of the most vascularized of all solid tumors and, therefore, represents an attractive target for antiangiogenic therapies. Many lesions, however, quickly develop escape mechanisms associated with changes in the tumor microenvironment (TME) resulting in rapid treatment failure. To prevent patients from adverse effects of ineffective therapy, there is a strong need to better predict and monitor antiangiogenic treatment response. PROCEDURES We utilized a novel physiological magnetic resonance imaging (MRI) method combining the visualization of oxygen metabolism and neovascularization for classification of five different TME compartments: necrosis, hypoxia with/without neovascularization, oxidative phosphorylation, and aerobic glycolysis. This approach, termed TME mapping, was used to monitor changes in tumor biology and pathophysiology within the TME in response to bevacizumab treatment in 18 patients with recurrent GB. RESULTS We detected dramatic changes in the TME by rearrangement of its compartments after the onset of bevacizumab treatment. All patients showed a decrease in active tumor volume and neovascularization as well as an increase in hypoxia and necrosis in the first follow-up after 3 months. We found that recurrent GB with a high percentage of neovascularization and active tumor before bevacizumab onset showed a poor or no treatment response. CONCLUSIONS TME mapping might be useful to develop strategies for patient stratification and response prediction before bevacizumab onset.
Collapse
|
7
|
Hvinden IC, Berg HE, Sachse D, Skaga E, Skottvoll FS, Lundanes E, Sandberg CJ, Vik-Mo EO, Rise F, Wilson SR. Nuclear Magnetic Resonance Spectroscopy to Identify Metabolite Biomarkers of Nonresponsiveness to Targeted Therapy in Glioblastoma Tumor Stem Cells. J Proteome Res 2019; 18:2012-2020. [PMID: 30964684 DOI: 10.1021/acs.jproteome.8b00801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Glioblastoma is the most common and malignant brain tumor, and current therapies confer only modest survival benefits. A major obstacle is our ability to monitor treatment effect on tumors. Current imaging modalities are ambiguous, and repeated biopsies are not encouraged. To scout for markers of treatment response, we used NMR spectroscopy to study the effects of a survivin inhibitor on the metabolome of primary glioblastoma cancer stem cells. Applying high resolution NMR spectroscopy (1H resonance frequency: 800.03 MHz) to just 3 million cells per sample, we achieved sensitive and high resolving determinations of, e.g., amino acids, nucleosides, and constituents of the citric acid cycle. For control samples that were cultured, prepared, and measured at varying dates, peak area relative standard deviations were 15-20%. Analyses of unfractionated lysates were performed for straightforward compound identification with COLMAR and HMDB databases. Principal component analysis revealed that citrate levels were clearly upregulated in nonresponsive cells, while lactate levels substantially decreased following treatment for both responsive and nonresponsive cells. Hence, lactate and citrate may be potential markers of successful drug uptake and poor response to survivin inhibitors, respectively. Our metabolomics approach provided alternative biomarker candidates compared to spectrometry-based proteomics, underlining benefits of complementary methodologies. These initial findings make a foundation for exploring in vivo MR spectroscopy (MRS) of brain tumors, as citrate and lactate are MRS-visible. In sum, NMR metabolomics is a tool for addressing glioblastoma.
Collapse
Affiliation(s)
- Ingvild Comfort Hvinden
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway.,Department of Chemistry , Chemistry Research Laboratory, University of Oxford , 12 Mansfield Road , Oxford OX1 3TA , United Kingdom
| | - Henriette Engen Berg
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway
| | - Daniel Sachse
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway
| | - Erlend Skaga
- Vilhelm Magnus Laboratory of Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery , Oslo University Hospital , 4950 Nydalen NO-0424 , Oslo , Norway.,Institute of Clinical Medicine, Faculty of Medicine , University of Oslo , Post Box 1171, Blindern NO-0318 , Oslo , Norway
| | - Frøydis Sved Skottvoll
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway.,Hybrid Technology Hub, Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine , University of Oslo , PO Box 1112, Blindern NO-0317 , Oslo , Norway
| | - Elsa Lundanes
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway
| | - Cecilie J Sandberg
- Vilhelm Magnus Laboratory of Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery , Oslo University Hospital , 4950 Nydalen NO-0424 , Oslo , Norway
| | - Einar O Vik-Mo
- Vilhelm Magnus Laboratory of Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery , Oslo University Hospital , 4950 Nydalen NO-0424 , Oslo , Norway
| | - Frode Rise
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway
| | - Steven Ray Wilson
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway.,Hybrid Technology Hub, Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine , University of Oslo , PO Box 1112, Blindern NO-0317 , Oslo , Norway
| |
Collapse
|
8
|
Zhu L, Wang J, Shi H, Tao X. Multimodality fMRI with perfusion, diffusion-weighted MRI and 1 H-MRS in the diagnosis of lympho-associated benign and malignant lesions of the parotid gland. J Magn Reson Imaging 2018; 49:423-432. [PMID: 30475438 DOI: 10.1002/jmri.26260] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Differential diagnosis of the mucosa-associated lymphoid tissue lymphoma (MALToma) and tumor-like benign lymphoepithelial lesion (BLEL) in the parotid gland is difficult. PURPOSE To distinguish MALToma and BLEL with multimodality MRI including hydrogenproton magnetic resonance spectroscopy (1 H-MRS), diffusion-weighted imaging (DWI-MR), and dynamic contrast-enhanced (DCE-MR), and evaluate each sequence. STUDY TYPE Retrospective. POPULATION Twenty-five patients with parotid tumor-like BLEL and 20 with parotid MALToma. FIELD STRENGTH/SEQUENCE 1.5-T/T1 WI, T2 WI, single-voxel 1 H-MRS, DWI-MR, and DCE-MR. ASSESSMENT All MR images were interpreted and agreed upon by two radiologists who were blinded to clinical information and histopathologic results. The imaging diagnoses were then compared to the histopathologic results. STATISTICAL TESTS Youden index was used to determine the optimized threshold value. The receiver operating characteristic (ROC) curve was used to evaluate the diagnostic efficiency of different functional (f)MRI methods. RESULTS Fisher's exact test indicated a significant difference between the 1 H-MRS images of the two lesions (P < 0.001). The sensitivity, specificity, and accuracy of positive choline (Cho) peak in 1 H-MRS of parotid MALToma were 80%, 76%, and 77.7%, respectively. The mean apparent diffusion coefficient (ADC) was 0.992 × 10-3 mm2 /s in patients with parotid tumor-like BLEL and 0.634 × 10-3 mm2 /s in patients with parotid MALToma, and the difference was statistically significant (t-test, P < 0.001). Choosing the Youden index as 0.669 × 10-3 mm2 /s, the sensitivity, specificity, and accuracy of the assay were 78.9%, 95.8%, and 88.4%, respectively. Assuming that time-intensity curve (TIC) type I indicated parotid MALToma (positive), and type II and type III indicated parotid tumor-like BLEL (negative), the sensitivity, specificity, and accuracy of time-to-peak (TTP) and initial slope of increase (ISI) in diagnosing MALToma were 94.1%, 95.2%, and 94.7%, respectively. Combining methods of TTP, ADC, and Cho peak reached the highest AUC (1.000). DATA CONCLUSION Combined use 1 H-MRS, DWI-MR, and DCE-MR increased the accuracy of the differential diagnosis between these lesions to 100%. Cho peak in 1 H-MRS, ADC less than 0.669 × 10-3 mm2 /s, TIC type I together indicated parotid MALToma. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2019;49:423-432.
Collapse
Affiliation(s)
- Ling Zhu
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Department of Radiology, Shanghai, P.R. China
| | - Jingbo Wang
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Department of Radiology, Shanghai, P.R. China
| | - Huimin Shi
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Department of Radiology, Shanghai, P.R. China
| | - Xiaofeng Tao
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Department of Radiology, Shanghai, P.R. China
| |
Collapse
|
9
|
Lenting K, Khurshed M, Peeters TH, van den Heuvel CNAM, van Lith SAM, de Bitter T, Hendriks W, Span PN, Molenaar RJ, Botman D, Verrijp K, Heerschap A, Ter Laan M, Kusters B, van Ewijk A, Huynen MA, van Noorden CJF, Leenders WPJ. Isocitrate dehydrogenase 1-mutated human gliomas depend on lactate and glutamate to alleviate metabolic stress. FASEB J 2018; 33:557-571. [PMID: 30001166 DOI: 10.1096/fj.201800907rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diffuse gliomas often carry point mutations in isocitrate dehydrogenase ( IDH1mut), resulting in metabolic stress. Although IDHmut gliomas are difficult to culture in vitro, they thrive in the brain via diffuse infiltration, suggesting brain-specific tumor-stroma interactions that can compensate for IDH-1 deficits. To elucidate the metabolic adjustments in clinical IDHmut gliomas that contribute to their malignancy, we applied a recently developed method of targeted quantitative RNA next-generation sequencing to 66 clinical gliomas and relevant orthotopic glioma xenografts, with and without the endogenous IDH-1R132H mutation. Datasets were analyzed in R using Manhattan plots to calculate distance between expression profiles, Ward's method to perform unsupervised agglomerative clustering, and the Mann Whitney U test and Fisher's exact tests for supervised group analyses. The significance of transcriptome data was investigated by protein analysis, in situ enzymatic activity mapping, and in vivo magnetic resonance spectroscopy of orthotopic IDH1mut- and IDHwt-glioma xenografts. Gene set enrichment analyses of clinical IDH1mut gliomas strongly suggest a role for catabolism of lactate and the neurotransmitter glutamate, whereas, in IDHwt gliomas, processing of glucose and glutamine are the predominant metabolic pathways. Further evidence of the differential metabolic activity in these cancers comes from in situ enzymatic mapping studies and preclinical in vivo magnetic resonance spectroscopy imaging. Our data support an evolutionary model in which IDHmut glioma cells exist in symbiosis with supportive neuronal cells and astrocytes as suppliers of glutamate and lactate, possibly explaining the diffuse nature of these cancers. The dependency on glutamate and lactate opens the way for novel approaches in the treatment of IDHmut gliomas.-Lenting, K., Khurshed, M., Peeters, T. H., van den Heuvel, C. N. A. M., van Lith, S. A. M., de Bitter, T., Hendriks, W., Span, P. N., Molenaar, R. J., Botman, D., Verrijp, K., Heerschap, A., ter Laan, M., Kusters, B., van Ewijk, A., Huynen, M. A., van Noorden, C. J. F., Leenders, W. P. J. Isocitrate dehydrogenase 1-mutated human gliomas depend on lactate and glutamate to alleviate metabolic stress.
Collapse
Affiliation(s)
- Krissie Lenting
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mohammed Khurshed
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Tom H Peeters
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Corina N A M van den Heuvel
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanne A M van Lith
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tessa de Bitter
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wiljan Hendriks
- Department of Cell Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Paul N Span
- Radiotherapy and Oncoimmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Remco J Molenaar
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Dennis Botman
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands; and
| | - Benno Kusters
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anne van Ewijk
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Martijn A Huynen
- Center for Molecular and Biomolecular Informatics, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Cornelis J F van Noorden
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - William P J Leenders
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
10
|
Khurshed M, Molenaar RJ, Lenting K, Leenders WP, van Noorden CJF. In silico gene expression analysis reveals glycolysis and acetate anaplerosis in IDH1 wild-type glioma and lactate and glutamate anaplerosis in IDH1-mutated glioma. Oncotarget 2018; 8:49165-49177. [PMID: 28467784 PMCID: PMC5564758 DOI: 10.18632/oncotarget.17106] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 04/03/2017] [Indexed: 12/15/2022] Open
Abstract
Hotspot mutations in isocitrate dehydrogenase 1 (IDH1) initiate low-grade glioma and secondary glioblastoma and induce a neomorphic activity that converts α-ketoglutarate (α-KG) to the oncometabolite D-2-hydroxyglutarate (D-2-HG). It causes metabolic rewiring that is not fully understood. We investigated the effects of IDH1 mutations (IDH1MUT) on expression of genes that encode for metabolic enzymes by data mining The Cancer Genome Atlas. We analyzed 112 IDH1 wild-type (IDH1WT) versus 399 IDH1MUT low-grade glioma and 157 IDH1WT versus 9 IDH1MUT glioblastoma samples. In both glioma types, IDH1WT was associated with high expression levels of genes encoding enzymes that are involved in glycolysis and acetate anaplerosis, whereas IDH1MUT glioma overexpress genes encoding enzymes that are involved in the oxidative tricarboxylic acid (TCA) cycle. In vitro, we observed that IDH1MUT cancer cells have a higher basal respiration compared to IDH1WT cancer cells and inhibition of the IDH1MUT shifts the metabolism by decreasing oxygen consumption and increasing glycolysis. Our findings indicate that IDH1WT glioma have a typical Warburg phenotype whereas in IDH1MUT glioma the TCA cycle, rather than glycolytic lactate production, is the predominant metabolic pathway. Our data further suggest that the TCA in IDH1MUT glioma is driven by lactate and glutamate anaplerosis to facilitate production of α-KG, and ultimately D-2-HG. This metabolic rewiring may be a basis for novel therapies for IDH1MUT and IDH1WT glioma.
Collapse
Affiliation(s)
- Mohammed Khurshed
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Remco J Molenaar
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Krissie Lenting
- Department of Pathology, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | | | - Cornelis J F van Noorden
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
11
|
Margiewicz S, Cordova C, Chi AS, Jain R. State of the Art Treatment and Surveillance Imaging of Glioblastomas. Semin Roentgenol 2017; 53:23-36. [PMID: 29405952 DOI: 10.1053/j.ro.2017.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
| | - Christine Cordova
- Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY
| | - Andrew S Chi
- Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY
| | - Rajan Jain
- Department of Radiology, NYU School of Medicine, New York, NY; Department of Neurosurgery, NYU School of Medicine, New York, NY.
| |
Collapse
|
12
|
Profiling of the metabolic transcriptome via single molecule molecular inversion probes. Sci Rep 2017; 7:11402. [PMID: 28900252 PMCID: PMC5595890 DOI: 10.1038/s41598-017-11035-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/18/2017] [Indexed: 12/20/2022] Open
Abstract
Cancer-specific metabolic alterations are of high interest as therapeutic targets. These alterations vary between tumor types, and to employ metabolic targeting to its fullest potential there is a need for robust methods that identify candidate targetable metabolic pathways in individual cancers. Currently, such methods include 13C-tracing studies and mass spectrometry/ magnetic resonance spectroscopic imaging. Due to high cost and complexity, such studies are restricted to a research setting. We here present the validation of a novel technique of metabolic profiling, based on multiplex targeted next generation sequencing of RNA with single molecule molecular inversion probes (smMIPs), designed to measure activity of and mutations in genes that encode metabolic enzymes. We here profiled an isogenic pair of cell lines, differing in expression of the Von Hippel Lindau protein, an important regulator of hypoxia-inducible genes. We show that smMIP-profiling provides relevant information on active metabolic pathways. Because smMIP-based targeted RNAseq is cost-effective and can be applied in a medium high-throughput setting (200 samples can be profiled simultaneously in one next generation sequencing run) it is a highly interesting approach for profiling of the activity of genes of interest, including those regulating metabolism, in a routine patient care setting.
Collapse
|
13
|
van den Heuvel CNAM, Navis AC, de Bitter T, Amiri H, Verrijp K, Heerschap A, Rex K, Dussault I, Caenepeel S, Coxon A, Span PN, Wesseling P, Hendriks W, Leenders WPJ. Selective MET Kinase Inhibition in MET-Dependent Glioma Models Alters Gene Expression and Induces Tumor Plasticity. Mol Cancer Res 2017; 15:1587-1597. [PMID: 28751462 DOI: 10.1158/1541-7786.mcr-17-0177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/15/2017] [Accepted: 07/24/2017] [Indexed: 11/16/2022]
Abstract
The receptor tyrosine kinase (RTK) MET represents a promising tumor target in a subset of glioblastomas. Most RTK inhibitors available in the clinic today, including those inhibiting MET, affect multiple targets simultaneously. Previously, it was demonstrated that treatment with cabozantinib (MET/VEGFR2/RET inhibitor) prolonged survival of mice carrying orthotopic patient-derived xenografts (PDX) of the MET-addicted glioblastoma model E98, yet did not prevent development of recurrent and cabozantinib-resistant tumors. To exclude VEGFR2 inhibition-inflicted blood-brain barrier normalization and diminished tumor distribution of the drug, we have now investigated the effects of the novel MET-selective inhibitor Compound A in the orthotopic E98 xenograft model. In vitro, Compound A proved a highly potent inhibitor of proliferation of MET-addicted cell lines. In line with its target selectivity, Compound A did not restore the leaky blood-brain barrier and was more effective than cabozantinib in inhibiting MET phosphorylation in vivo Compound A treatment significantly prolonged survival of mice carrying E98 tumor xenografts, but did not prevent eventual progression. Contrasting in vitro results, the Compound A-treated xenografts displayed high levels of AKT phosphorylation despite the absence of phosphorylated MET. Profiling by RNA sequencing showed that in vivo transcriptomes differed significantly from those in control xenografts.Implications: Collectively, these findings demonstrate the plasticity of paracrine growth factor receptor signaling in vivo and urge for prudency with in vitro drug-testing strategies to validate monotherapies. Mol Cancer Res; 15(11); 1587-97. ©2017 AACR.
Collapse
Affiliation(s)
| | - Anna C Navis
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Tessa de Bitter
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Houshang Amiri
- Department of Radiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Arend Heerschap
- Department of Radiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Karen Rex
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Isabelle Dussault
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Sean Caenepeel
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Angela Coxon
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Paul N Span
- Department of Radiation Oncology, Radboud University Medical Centre, Radiotherapy and Oncoimmunology Laboratory, Nijmegen, the Netherlands
| | - Pieter Wesseling
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Wiljan Hendriks
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - William P J Leenders
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands.
| |
Collapse
|
14
|
Evaluation of human glioma using in-vivo proton magnetic resonance spectroscopy combined with expression of cyclooxygenase-2: a preliminary clinical trial. Neuroreport 2017; 28:414-420. [PMID: 28306608 DOI: 10.1097/wnr.0000000000000771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We sought to investigate the correlation between in-vivo proton magnetic resonance spectroscopy (H-MRS) and cyclooxygenase-2 (COX-2) expression in human glioma, and to advance their roles in diagnostic mapping and monitoring of glioma biological behavior. Thirty-nine patients with different grades of glioma (WHO classification I-IV) included in this study were scanned at 3.0 T MR before operation or biopsy puncture. Tumor morphology and H-MRS metabolites ratio [choline (Cho)/creatine (Cr)] were evaluated independently by two experienced radiologists. Paraffin-embedded glioma specimens were detected for the COX-2 expression using immunohistochemistry. Parametric and nonparametric tests including correlation, logistic regression and receiver operating characteristic analysis were applied to assess the predicative roles and relation diagram of H-MRS and COX-2. The COX-2 expression showed a significant difference between low and high grade gliomas (P<0.01). Varying degrees of COX-2 expression have positive correlation with the Cho/Cr values in tumor zone (r=0.49, P=0.013), and showed not significant correlation with sex, age, and tumor location. For patients with high grade gliomas after surgery and radiation, COX-2 was associated with shortened survival in univariate analysis (P=0.025). The COX-2, Cho/Cr value and age were the significant prognostic indicators shown in multivariate survival analysis. The COX-2 and Cho/Cr value of H-MRS have correlation, and are both positive indicators for overall survival of human high grade glioma, and could be combined as a joint role to provide more evidences to assess the biological behavior.
Collapse
|
15
|
Mahase S, Rattenni RN, Wesseling P, Leenders W, Baldotto C, Jain R, Zagzag D. Hypoxia-Mediated Mechanisms Associated with Antiangiogenic Treatment Resistance in Glioblastomas. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:940-953. [PMID: 28284719 PMCID: PMC5417003 DOI: 10.1016/j.ajpath.2017.01.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/31/2016] [Accepted: 01/05/2017] [Indexed: 12/28/2022]
Abstract
Glioblastomas (GBMs) are malignant tumors characterized by their vascularity and invasive capabilities. Antiangiogenic therapy (AAT) is a treatment option that targets GBM-associated vasculature to mitigate the growth of GBMs. However, AAT demonstrates transient effects because many patients eventually develop resistance to this treatment. Several recent studies attempt to explain the molecular and biochemical basis of resistance to AAT in GBM patients. Experimental investigations suggest that the induction of extensive intratumoral hypoxia plays a key role in GBM escape from AAT. In this review, we examine AAT resistance in GBMs, with an emphasis on six potential hypoxia-mediated mechanisms: enhanced invasion and migration, including increased expression of matrix metalloproteinases and activation of the c-MET tyrosine kinase pathway; shifts in cellular metabolism, including up-regulation of hypoxia inducible factor-1α's downstream processes and the Warburg effect; induction of autophagy; augmentation of GBM stem cell self-renewal; possible implications of GBM-endothelial cell transdifferentiation; and vasoformative responses, including vasculogenesis, alternative angiogenic pathways, and vascular mimicry. Juxtaposing recent studies on well-established resistance pathways with that of emerging mechanisms highlights the overall complexity of GBM treatment resistance while also providing direction for further investigation.
Collapse
Affiliation(s)
- Sean Mahase
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York
| | - Rachel N Rattenni
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands; Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center, Utrecht, the Netherlands
| | - William Leenders
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clarissa Baldotto
- Medical Oncology, Instituto Nacionale de Cancer, Rio de Janeiro, Brazil
| | - Rajan Jain
- Department of Radiology, New York University School of Medicine, New York, New York; Department of Neurosurgery, New York University School of Medicine, New York, New York
| | - David Zagzag
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York; Department of Neurosurgery, New York University School of Medicine, New York, New York; Division of Neuropathology, Department of Pathology, New York University School of Medicine, New York, New York; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, New York.
| |
Collapse
|
16
|
Lenting K, Verhaak R, Ter Laan M, Wesseling P, Leenders W. Glioma: experimental models and reality. Acta Neuropathol 2017; 133:263-282. [PMID: 28074274 PMCID: PMC5250671 DOI: 10.1007/s00401-017-1671-4] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/12/2022]
Abstract
In theory, in vitro and in vivo models for human gliomas have great potential to not only enhance our understanding of glioma biology, but also to facilitate the development of novel treatment strategies for these tumors. For reliable prediction and validation of the effects of different therapeutic modalities, however, glioma models need to comply with specific and more strict demands than other models of cancer, and these demands are directly related to the combination of genetic aberrations and the specific brain micro-environment gliomas grow in. This review starts with a brief introduction on the pathological and molecular characteristics of gliomas, followed by an overview of the models that have been used in the last decades in glioma research. Next, we will discuss how these models may play a role in better understanding glioma development and especially in how they can aid in the design and optimization of novel therapies. The strengths and weaknesses of the different models will be discussed in light of genotypic, phenotypic and metabolic characteristics of human gliomas. The last part of this review provides some examples of how therapy experiments using glioma models can lead to deceptive results when such characteristics are not properly taken into account.
Collapse
Affiliation(s)
- Krissie Lenting
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Roel Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, Utrecht, The Netherlands
| | - William Leenders
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
17
|
van Lith SAM, Navis AC, Lenting K, Verrijp K, Schepens JTG, Hendriks WJAJ, Schubert NA, Venselaar H, Wevers RA, van Rooij A, Wesseling P, Molenaar RJ, van Noorden CJF, Pusch S, Tops B, Leenders WPJ. Identification of a novel inactivating mutation in Isocitrate Dehydrogenase 1 (IDH1-R314C) in a high grade astrocytoma. Sci Rep 2016; 6:30486. [PMID: 27460417 PMCID: PMC4962051 DOI: 10.1038/srep30486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022] Open
Abstract
The majority of low-grade and secondary high-grade gliomas carry heterozygous hotspot mutations in cytosolic isocitrate dehydrogenase 1 (IDH1) or the mitochondrial variant IDH2. These mutations mostly involve Arg132 in IDH1, and Arg172 or Arg140 in IDH2. Whereas IDHs convert isocitrate to alpha-ketoglutarate (α-KG) with simultaneous reduction of NADP+ to NADPH, these IDH mutants reduce α-KG to D-2-hydroxyglutarate (D-2-HG) while oxidizing NADPH. D-2-HG is a proposed oncometabolite, acting via competitive inhibition of α-KG-dependent enzymes that are involved in metabolism and epigenetic regulation. However, much less is known about the implications of the metabolic stress, imposed by decreased α-KG and NADPH production, for tumor biology. We here present a novel heterozygous IDH1 mutation, IDH1R314C, which was identified by targeted next generation sequencing of a high grade glioma from which a mouse xenograft model and a cell line were generated. IDH1R314C lacks isocitrate-to-α-KG conversion activity due to reduced affinity for NADP+, and differs from the IDH1R132 mutants in that it does not produce D-2-HG. Because IDH1R314C is defective in producing α-KG and NADPH, without concomitant production of the D-2-HG, it represents a valuable tool to study the effects of IDH1-dysfunction on cellular metabolism in the absence of this oncometabolite.
Collapse
Affiliation(s)
| | - Anna C Navis
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Krissie Lenting
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Jan T G Schepens
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Wiljan J A J Hendriks
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Nil A Schubert
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Hanka Venselaar
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Arno van Rooij
- Translational Metabolic Laboratory, Department Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands.,Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Remco J Molenaar
- Department of Cell Biology and Histology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Stefan Pusch
- Clinical Cooperation Unit Neuropathology, German Cancer Center (DKFZ), Heidelberg, Germany
| | - Bastiaan Tops
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | | |
Collapse
|
18
|
Delgado-Goñi T, Ortega-Martorell S, Ciezka M, Olier I, Candiota AP, Julià-Sapé M, Fernández F, Pumarola M, Lisboa PJ, Arús C. MRSI-based molecular imaging of therapy response to temozolomide in preclinical glioblastoma using source analysis. NMR IN BIOMEDICINE 2016; 29:732-743. [PMID: 27061401 DOI: 10.1002/nbm.3521] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 02/14/2016] [Accepted: 02/23/2016] [Indexed: 06/05/2023]
Abstract
Characterization of glioblastoma (GB) response to treatment is a key factor for improving patients' survival and prognosis. MRI and magnetic resonance spectroscopic imaging (MRSI) provide morphologic and metabolic profiles of GB but usually fail to produce unequivocal biomarkers of response. The purpose of this work is to provide proof of concept of the ability of a semi-supervised signal source extraction methodology to produce images with robust recognition of response to temozolomide (TMZ) in a preclinical GB model. A total of 38 female C57BL/6 mice were used in this study. The semi-supervised methodology extracted the required sources from a training set consisting of MRSI grids from eight GL261 GBs treated with TMZ, and six control untreated GBs. Three different sources (normal brain parenchyma, actively proliferating GB and GB responding to treatment) were extracted and used for calculating nosologic maps representing the spatial response to treatment. These results were validated with an independent test set (7 control and 17 treated cases) and correlated with histopathology. Major differences between the responder and non-responder sources were mainly related to the resonances of mobile lipids (MLs) and polyunsaturated fatty acids in MLs (0.9, 1.3 and 2.8 ppm). Responding tumors showed significantly lower mitotic (3.3 ± 2.9 versus 14.1 ± 4.2 mitoses/field) and proliferation rates (29.8 ± 10.3 versus 57.8 ± 5.4%) than control untreated cases. The methodology described in this work is able to produce nosological images of response to TMZ in GL261 preclinical GBs and suitably correlates with the histopathological analysis of tumors. A similar strategy could be devised for monitoring response to treatment in patients. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- T Delgado-Goñi
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | - S Ortega-Martorell
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Department of Mathematics and Statistics, Liverpool John Moores University, Liverpool, UK
| | - M Ciezka
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - I Olier
- Institute for Science and Technology in Medicine, Keele University, Stoke-On-Trent, UK
- Centre for Health Informatics, Institute of Population Health University of Manchester, Manchester, UK
| | - A P Candiota
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - M Julià-Sapé
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - F Fernández
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Departament de Medicina i Cirurgia Animals, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - M Pumarola
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Departament de Medicina i Cirurgia Animals, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - P J Lisboa
- Department of Mathematics and Statistics, Liverpool John Moores University, Liverpool, UK
| | - C Arús
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
19
|
Harris LM, Tunariu N, Messiou C, Hughes J, Wallace T, DeSouza NM, Leach MO, Payne GS. Evaluation of lactate detection using selective multiple quantum coherence in phantoms and brain tumours. NMR IN BIOMEDICINE 2015; 28:338-43. [PMID: 25586623 PMCID: PMC4681317 DOI: 10.1002/nbm.3255] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 05/28/2023]
Abstract
Lactate is a product of glucose metabolism. In tumour tissues, which exhibit enhanced glycolytic metabolism, lactate signals may be elevated, making lactate a potential useful tumour biomarker. Methods of lactate quantitation are complicated because of overlap between the lactate methyl doublet CH3 resonance and a lipid resonance at 1.3 ppm. This study presents the use of a selective homonuclear multiple quantum coherence transfer sequence (SelMQC-CSI), at 1.5 T, to better quantify lactate in the presence of lipids. Work performed on phantoms showed good lactate detection (49%) and lipid suppression (98%) efficiencies. To evaluate the method in the brain, the sequence was tested on a group of 23 patients with treated brain tumours, either glioma (N=20) or secondary metastases in the brain (N=3). Here it was proved to be of use in determining lactate concentrations in vivo. Lactate was clearly seen in SelMQC spectra of glioma, even in the presence of lipids, with high grade glioma (7.3 ± 1.9 mM, mean ± standard deviation) having higher concentrations than low grade glioma (1.9 ± 1.5 mM, p=0.048). Lactate was not seen in secondary metastases in the brain. SelMQC-CSI is shown to be a useful technique for measuring lactate in tumours whose signals are otherwise contaminated by lipid.
Collapse
Affiliation(s)
- L M Harris
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research and Royal Marsden NHS Foundation TrustSutton, Surrey, UK
| | - N Tunariu
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research and Royal Marsden NHS Foundation TrustSutton, Surrey, UK
| | - C Messiou
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research and Royal Marsden NHS Foundation TrustSutton, Surrey, UK
| | - J Hughes
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research and Royal Marsden NHS Foundation TrustSutton, Surrey, UK
| | - T Wallace
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research and Royal Marsden NHS Foundation TrustSutton, Surrey, UK
| | - N M DeSouza
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research and Royal Marsden NHS Foundation TrustSutton, Surrey, UK
| | - M O Leach
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research and Royal Marsden NHS Foundation TrustSutton, Surrey, UK
| | - G S Payne
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research and Royal Marsden NHS Foundation TrustSutton, Surrey, UK
| |
Collapse
|
20
|
Fack F, Espedal H, Keunen O, Golebiewska A, Obad N, Harter PN, Mittelbronn M, Bähr O, Weyerbrock A, Stuhr L, Miletic H, Sakariassen PØ, Stieber D, Rygh CB, Lund-Johansen M, Zheng L, Gottlieb E, Niclou SP, Bjerkvig R. Bevacizumab treatment induces metabolic adaptation toward anaerobic metabolism in glioblastomas. Acta Neuropathol 2015; 129:115-31. [PMID: 25322816 PMCID: PMC4282692 DOI: 10.1007/s00401-014-1352-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 09/30/2014] [Accepted: 10/01/2014] [Indexed: 10/29/2022]
Abstract
Anti-angiogenic therapy in glioblastoma (GBM) has unfortunately not led to the anticipated improvement in patient prognosis. We here describe how human GBM adapts to bevacizumab treatment at the metabolic level. By performing (13)C6-glucose metabolic flux analysis, we show for the first time that the tumors undergo metabolic re-programming toward anaerobic metabolism, thereby uncoupling glycolysis from oxidative phosphorylation. Following treatment, an increased influx of (13)C6-glucose was observed into the tumors, concomitant to increased lactate levels and a reduction of metabolites associated with the tricarboxylic acid cycle. This was confirmed by increased expression of glycolytic enzymes including pyruvate dehydrogenase kinase in the treated tumors. Interestingly, L-glutamine levels were also reduced. These results were further confirmed by the assessment of in vivo metabolic data obtained by magnetic resonance spectroscopy and positron emission tomography. Moreover, bevacizumab led to a depletion in glutathione levels indicating that the treatment caused oxidative stress in the tumors. Confirming the metabolic flux results, immunohistochemical analysis showed an up-regulation of lactate dehydrogenase in the bevacizumab-treated tumor core as well as in single tumor cells infiltrating the brain, which may explain the increased invasion observed after bevacizumab treatment. These observations were further validated in a panel of eight human GBM patients in which paired biopsy samples were obtained before and after bevacizumab treatment. Importantly, we show that the GBM adaptation to bevacizumab therapy is not mediated by clonal selection mechanisms, but represents an adaptive response to therapy.
Collapse
Affiliation(s)
- Fred Fack
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Centre de Recherche Public de la Santé, Strassen, Luxembourg
| | - Heidi Espedal
- NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5019 Bergen, Norway
| | - Olivier Keunen
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Centre de Recherche Public de la Santé, Strassen, Luxembourg
| | - Anna Golebiewska
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Centre de Recherche Public de la Santé, Strassen, Luxembourg
| | - Nina Obad
- NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5019 Bergen, Norway
| | - Patrick N. Harter
- Edinger Institute, Institute of Neurology, Goethe University, Hospital Frankfurt, Frankfurt am Main, Germany
| | - Michel Mittelbronn
- Edinger Institute, Institute of Neurology, Goethe University, Hospital Frankfurt, Frankfurt am Main, Germany
| | - Oliver Bähr
- Dr. Senckenberg Institute of Neurooncology, Goethe University, Hospital Frankfurt, Frankfurt am Main, Germany
| | - Astrid Weyerbrock
- Department of Neurosurgery, University Hospital Freiburg, Freiburg, Germany
| | - Linda Stuhr
- Matrix Biology Group, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Hrvoje Miletic
- NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5019 Bergen, Norway
- Department of Pathology, Haukeland University Hospital, The Gade Institute, Bergen, Norway
- KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway
| | - Per Ø. Sakariassen
- NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5019 Bergen, Norway
| | - Daniel Stieber
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Centre de Recherche Public de la Santé, Strassen, Luxembourg
| | - Cecilie B. Rygh
- Department of Biomedicine, Molecular Imaging Center, University of Bergen, Bergen, Norway
| | - Morten Lund-Johansen
- Department of Neurosurgery, Haukeland University Hospital, Bergen, Norway
- KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway
| | - Liang Zheng
- Cancer Research UK, Beatson Institute, Glasgow, Scotland, UK
| | - Eyal Gottlieb
- Cancer Research UK, Beatson Institute, Glasgow, Scotland, UK
| | - Simone P. Niclou
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Centre de Recherche Public de la Santé, Strassen, Luxembourg
- KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway
| | - Rolf Bjerkvig
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Centre de Recherche Public de la Santé, Strassen, Luxembourg
- NorLux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5019 Bergen, Norway
- KG Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway
| |
Collapse
|
21
|
Delgado-Goñi T, Julià-Sapé M, Candiota AP, Pumarola M, Arús C. Molecular imaging coupled to pattern recognition distinguishes response to temozolomide in preclinical glioblastoma. NMR IN BIOMEDICINE 2014; 27:1333-1345. [PMID: 25208348 DOI: 10.1002/nbm.3194] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 07/24/2014] [Accepted: 07/27/2014] [Indexed: 06/03/2023]
Abstract
Non-invasive monitoring of response to treatment of glioblastoma (GB) is nowadays carried out using MRI. MRS and MR spectroscopic imaging (MRSI) constitute promising tools for this undertaking. A temozolomide (TMZ) protocol was optimized for GL261 GB. Sixty-three mice were studied by MRI/MRS/MRSI. The spectroscopic information was used for the classification of control brain and untreated and responding GB, and validated against post-mortem immunostainings in selected animals. A classification system was developed, based on the MRSI-sampled metabolome of normal brain parenchyma, untreated and responding GB, with a 93% accuracy. Classification of an independent test set yielded a balanced error rate of 6% or less. Classifications correlated well both with tumor volume changes detected by MRI after two TMZ cycles and with the histopathological data: a significant decrease (p < 0.05) in the proliferation and mitotic rates and a 4.6-fold increase in the apoptotic rate. A surrogate response biomarker based on the linear combination of 12 spectral features has been found in the MRS/MRSI pattern of treated tumors, allowing the non-invasive classification of growing and responding GL261 GB. The methodology described can be applied to preclinical treatment efficacy studies to test new antitumoral drugs, and begets translational potential for early response detection in clinical studies.
Collapse
Affiliation(s)
- Teresa Delgado-Goñi
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey, SM2 5PT, UK
| | | | | | | | | |
Collapse
|
22
|
Effect of acute hyperglycemia on moderately hypothermic GL261 mouse glioma monitored by T1-weighted DCE MRI. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2014; 28:119-26. [PMID: 24916487 DOI: 10.1007/s10334-014-0447-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 04/15/2014] [Accepted: 04/15/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE We sought to evaluate the effects of acute hyperglycemia induced by intraperitoneal injection of glucose (2.7 g/kg) on vascular delivery to GL261 mouse gliomas kept at moderate hypothermia (~30 °C). MATERIALS AND METHODS Seven GL261 glioma-bearing mice were studied by T1-weighted DCE MRI before and after an injection of glucose (n = 4) or saline (n = 3). Maximum relative contrast enhancement (RCE) and initial area under the enhancement curve (IAUC) were determined in each pixel. RESULTS The mean tumor parameter values showed no significant changes after injecting either saline (RCE -5.9 ± 5.0 %; IAUC -3.7 ± 3.6 %) or glucose (RCE -1.6 ± 9.0 %; IAUC +0.6 ± 6.4 %). Pixel-by-pixel analysis revealed small post-injection changes in RCE and IAUC between the glucose and saline groups, all within 13 % range of their baseline values. CONCLUSION Perturbing the metabolism of GL261 tumors kept at moderate hypothermia with hyperglycemia did not induce significant changes in the permeability/perfusion of these tumors. This is relevant for future studies with this model since regional differences in glucose accumulation could thus reflect basal heterogeneities in vasculature and/or metabolism of GL261 tumors.
Collapse
|
23
|
Chronaiou I, Stensjøen AL, Sjøbakk TE, Esmaeili M, Bathen TF. Impacts of MR spectroscopic imaging on glioma patient management. Acta Oncol 2014; 53:580-9. [PMID: 24628262 DOI: 10.3109/0284186x.2014.891046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Magnetic resonance (MR) modalities are routine imaging tools in the diagnosis and management of gliomas. MR spectroscopic imaging (MRSI), which relies on the metabolic characteristics of tissues, has been developed to accelerate the understanding of gliomas and to aid in effective clinical decision making and development of targeted therapies. In this review, the potentials and practical challenges to frequently use this technique in clinical management of gliomas are discussed. The applications of new biomarkers detectable by MRSI in differential glioma diagnosis, pre- and post-treatment evaluations, and neurosurgery are also addressed.
Collapse
Affiliation(s)
- Ioanna Chronaiou
- Radiography Department, Faculty of Technology (AFT), Sør-Trøndelag University College (HiST) , Trondheim , Norway
| | | | | | | | | |
Collapse
|
24
|
Glutamate as chemotactic fuel for diffuse glioma cells: are they glutamate suckers? Biochim Biophys Acta Rev Cancer 2014; 1846:66-74. [PMID: 24747768 DOI: 10.1016/j.bbcan.2014.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/09/2014] [Accepted: 04/11/2014] [Indexed: 11/21/2022]
Abstract
Diffuse gliomas comprise a group of primary brain tumors that originate from glial (precursor) cells and present as a variety of malignancy grades which have in common that they grow by diffuse infiltration. This phenotype complicates treatment enormously as it precludes curative surgery and radiotherapy. Furthermore, diffusely infiltrating glioma cells often hide behind a functional blood-brain barrier, hampering delivery of systemically administered therapeutic and diagnostic compounds to the tumor cells. The present review addresses the biological mechanisms that underlie the diffuse infiltrative phenotype, knowledge of which may improve treatment strategies for this disastrous tumor type. The invasive phenotype is specific for glioma: most other brain tumor types, both primary and metastatic, grow as delineated lesions. Differences between the genetic make-up of glioma and that of other tumor types may therefore help to unravel molecular pathways, involved in diffuse infiltrative growth. One such difference concerns mutations in the NADP(+)-dependent isocitrate dehydrogenase (IDH1 and IDH2) genes, which occur in >80% of cases of low grade glioma and secondary glioblastoma. In this review we present a novel hypothesis which links IDH1 and IDH2 mutations to glutamate metabolism, possibly explaining the specific biological behavior of diffuse glioma.
Collapse
|
25
|
Pun WK, Chow SP, Fang D, Cheng CL, Leong JC, Ng C. Post-traumatic oedema of the foot after tibial fracture. Expert Rev Mol Diagn 1990; 15:735-47. [PMID: 2592102 DOI: 10.1586/14737159.2015.1039515] [Citation(s) in RCA: 88] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A total of 97 patients with diaphyseal tibial fractures treated with functional bracing were studied prospectively. Persistent ipsilateral foot swelling was present in 84.5 per cent of the patients. Most of the swellings subsided with time, but a small percentage of them persisted for a duration of 2 years or more after injury. The time for disappearance of the swelling in 50 per cent of the patients was 18.6 weeks. The development of oedema is not related to the age and sex of the patients, the configuration, type and level of the fractures, or the association of a fibular fracture. The bone healed quicker in those who did not have swelling of the foot. Once the swelling has developed, it seems to run its own course and its disappearance is not related to the age and sex, the configuration, type and level of fractures, the association of a fibular fracture, or the time for fracture healing. This complication does not have any adverse effect on the functional recovery of the patients.
Collapse
Affiliation(s)
- W K Pun
- Department of Orthopaedic Surgery, University of Hong Kong, Queen Mary Hospital
| | | | | | | | | | | |
Collapse
|