1
|
Godlewski A, Mojsak P, Pienkowski T, Lyson T, Mariak Z, Reszec J, Kaminski K, Moniuszko M, Kretowski A, Ciborowski M. Metabolomic profiling of plasma from glioma and meningioma patients based on two complementary mass spectrometry techniques. Metabolomics 2025; 21:33. [PMID: 39987409 DOI: 10.1007/s11306-025-02231-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/02/2025] [Indexed: 02/24/2025]
Abstract
INTRODUCTION Extracranial and intracranial tumors are a diverse group of malignant and benign neoplasms, influenced by multiple factors. Given the complex nature of these tumors and usually late or accidental diagnosis, minimally invasive, rapid, early, and accurate diagnostic methods are urgently required. Metabolomics offers promising insights into central nervous system tumors by uncovering distinctive metabolic changes linked to tumor development. OBJECTIVES This study aimed to elucidate the role of altered metabolites and the associated biological pathways implicated in the development of gliomas and meningiomas. METHODS The study was conducted on 95 patients with gliomas, 68 patients with meningiomas, and 71 subjects as a control group. The metabolic profiling of gliomas and meningiomas achieved by integrating untargeted metabolomic analysis based on GC-MS and targeted analysis performed using LC-MS/MS represents the first comprehensive study. Three comparisons (gliomas or meningiomas vs. controls as well as gliomas vs. meningiomas) were performed to reveal statistically significant metabolites. RESULTS Comparative analysis revealed 97, 56, and 27 significant metabolites for gliomas vs. controls, meningiomas vs. controls and gliomas vs. meningiomas comparison, respectively. Moreover, among above mentioned comparisons unique metabolites involved in arginine biosynthesis and metabolism, the Krebs cycle, and lysine degradation pathways were found. Notably, 2-aminoadipic acid has been identified as a metabolite that can be used in distinguishing two tumor types. CONCLUSIONS Our results provide a deeper understanding of the metabolic changes associated with brain tumor development and progression.
Collapse
Affiliation(s)
- Adrian Godlewski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Patrycja Mojsak
- Clinical Research Centre, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Tomasz Pienkowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Tomasz Lyson
- Department of Neurosurgery, Medical University of Bialystok, Bialystok, 15-276, Poland
- Department of Interventional Neurology, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Zenon Mariak
- Department of Neurosurgery, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Joanna Reszec
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Karol Kaminski
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, 15-276, Poland
- Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, 15-276, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, 15-276, Poland.
| |
Collapse
|
2
|
Wang N, Yuan Y, Hu T, Xu H, Piao H. Metabolism: an important player in glioma survival and development. Discov Oncol 2024; 15:577. [PMID: 39436434 PMCID: PMC11496451 DOI: 10.1007/s12672-024-01402-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
Gliomas are malignant tumors originating from both neuroglial cells and neural stem cells. The involvement of neural stem cells contributes to the tumor's heterogeneity, affecting its metabolic features, development, and response to therapy. This review provides a brief introduction to the importance of metabolism in gliomas before systematically categorizing them into specific groups based on their histological and molecular genetic markers. Metabolism plays a critical role in glioma biology, as tumor cells rely heavily on altered metabolic pathways to support their rapid growth, survival, and progression. Dysregulated metabolic processes, involving carbohydrates, lipids, and amino acids not only fuel tumor development but also contribute to therapy resistance and metastatic potential. By understanding these metabolic changes, key intervention points, such as mutations in genes like RTK, EGFR, RAS, and IDH can be identified, paving the way for novel therapeutic strategies. This review emphasizes the connection between metabolic pathways and clinical challenges, offering actionable insights for future research and therapeutic development in gliomas.
Collapse
Affiliation(s)
- Ning Wang
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Shenyang, Dadong, 110042, P R China
- Institute of Cancer Medicine, Dalian University of Technology, No.2 Linggong Road, Dalian, Ganjingzi, 116024, P R China
| | - Yiru Yuan
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Shenyang, Dadong, 110042, P R China
- Institute of Cancer Medicine, Dalian University of Technology, No.2 Linggong Road, Dalian, Ganjingzi, 116024, P R China
| | - Tianhao Hu
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Shenyang, Dadong, 110042, P R China
- Institute of Cancer Medicine, Dalian University of Technology, No.2 Linggong Road, Dalian, Ganjingzi, 116024, P R China
| | - Huizhe Xu
- Institute of Cancer Medicine, Dalian University of Technology, No.2 Linggong Road, Dalian, Ganjingzi, 116024, P R China.
- Central Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Shenyang, Liaoning Province, 110042, P R China.
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Shenyang, Dadong, 110042, P R China.
- Institute of Cancer Medicine, Dalian University of Technology, No.2 Linggong Road, Dalian, Ganjingzi, 116024, P R China.
| |
Collapse
|
3
|
Adamson PM, Datta K, Watkins R, Recht LD, Hurd RE, Spielman DM. Deuterium metabolic imaging for 3D mapping of glucose metabolism in humans with central nervous system lesions at 3T. Magn Reson Med 2024; 91:39-50. [PMID: 37796151 PMCID: PMC10841984 DOI: 10.1002/mrm.29830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/19/2023] [Accepted: 07/28/2023] [Indexed: 10/06/2023]
Abstract
PURPOSE To explore the potential of 3T deuterium metabolic imaging (DMI) using a birdcage 2 H radiofrequency (RF) coil in both healthy volunteers and patients with central nervous system (CNS) lesions. METHODS A modified gradient filter, home-built 2 H volume RF coil, and spherical k-space sampling were employed in a three-dimensional chemical shift imaging acquisition to obtain high-quality whole-brain metabolic images of 2 H-labeled water and glucose metabolic products. These images were acquired in a healthy volunteer and three subjects with CNS lesions of varying pathologies. Hardware and pulse sequence experiments were also conducted to improve the signal-to-noise ratio of DMI at 3T. RESULTS The ability to quantify local glucose metabolism in correspondence to anatomical landmarks across patients with varying CNS lesions is demonstrated, and increased lactate is observed in one patient with the most active disease. CONCLUSION DMI offers the potential to examine metabolic activity in human subjects with CNS lesions with DMI at 3T, promising for the potential of the future clinical translation of this metabolic imaging technique.
Collapse
Affiliation(s)
- Philip M. Adamson
- Department of Electrical Engineering, Stanford University, Stanford, California USA
| | - Keshav Datta
- Department of Radiology, Stanford University, Stanford, California, USA
| | - Ron Watkins
- Department of Radiology, Stanford University, Stanford, California, USA
| | - Lawrence D. Recht
- Department of Neurology, Stanford University, Stanford, California, USA
| | - Ralph E. Hurd
- Department of Radiology, Stanford University, Stanford, California, USA
| | | |
Collapse
|
4
|
Vassileva V, Braga M, Barnes C, Przystal J, Ashek A, Allott L, Brickute D, Abrahams J, Suwan K, Carcaboso AM, Hajitou A, Aboagye EO. Effective Detection and Monitoring of Glioma Using [ 18F]FPIA PET Imaging. Biomedicines 2021; 9:811. [PMID: 34356874 PMCID: PMC8301305 DOI: 10.3390/biomedicines9070811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Reprogrammed cellular metabolism is a cancer hallmark. In addition to increased glycolysis, the oxidation of acetate in the citric acid cycle is another common metabolic phenotype. We have recently developed a novel fluorine-18-labelled trimethylacetate-based radiotracer, [18F]fluoro-pivalic acid ([18F]FPIA), for imaging the transcellular flux of short-chain fatty acids, and investigated whether this radiotracer can be used for the detection of glioma growth. METHODS We evaluated the potential of [18F]FPIA PET to monitor tumor growth in orthotopic patient-derived (HSJD-GBM-001) and cell line-derived (U87, LN229) glioma xenografts, and also included [18F]FDG PET for comparison. We assessed proliferation (Ki-67) and the expression of lipid metabolism and transport proteins (CPT1, SLC22A2, SLC22A5, SLC25A20) by immunohistochemistry, along with etomoxir treatment to provide insights into [18F]FPIA uptake. RESULTS Longitudinal PET imaging showed gradual increase in [18F]FPIA uptake in orthotopic glioma models with disease progression (p < 0.0001), and high tumor-to-brain contrast compared to [18F]FDG (p < 0.0001). [18F]FPIA uptake correlated positively with Ki-67 (p < 0.01), SLC22A5 (p < 0.001) and SLC25A20 (p = 0.001), and negatively with CPT1 (p < 0.01) and SLC22A2 (p < 0.01). Etomoxir reduced [18F]FPIA uptake, which correlated with decreased Ki-67 (p < 0.05). CONCLUSIONS Our findings support the use of [18F]FPIA PET for the detection and longitudinal monitoring of glioma, showing a positive correlation with tumor proliferation, and suggest transcellular flux-mediated radiotracer uptake.
Collapse
Affiliation(s)
- Vessela Vassileva
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Marta Braga
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Chris Barnes
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Justyna Przystal
- Department of Medicine, Division of Brain Sciences, Imperial College London, Hammersmith Campus, Burlington Danes, London W12 0NN, UK; (J.P.); (K.S.); (A.H.)
| | - Ali Ashek
- Department of Medicine, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK;
| | - Louis Allott
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Diana Brickute
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Joel Abrahams
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Keittisak Suwan
- Department of Medicine, Division of Brain Sciences, Imperial College London, Hammersmith Campus, Burlington Danes, London W12 0NN, UK; (J.P.); (K.S.); (A.H.)
| | | | - Amin Hajitou
- Department of Medicine, Division of Brain Sciences, Imperial College London, Hammersmith Campus, Burlington Danes, London W12 0NN, UK; (J.P.); (K.S.); (A.H.)
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| |
Collapse
|
5
|
Cruz Da Silva E, Mercier MC, Etienne-Selloum N, Dontenwill M, Choulier L. A Systematic Review of Glioblastoma-Targeted Therapies in Phases II, III, IV Clinical Trials. Cancers (Basel) 2021; 13:1795. [PMID: 33918704 PMCID: PMC8069979 DOI: 10.3390/cancers13081795] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM), the most frequent and aggressive glial tumor, is currently treated as first line by the Stupp protocol, which combines, after surgery, radiotherapy and chemotherapy. For recurrent GBM, in absence of standard treatment or available clinical trials, various protocols including cytotoxic drugs and/or bevacizumab are currently applied. Despite these heavy treatments, the mean overall survival of patients is under 18 months. Many clinical studies are underway. Based on clinicaltrials.org and conducted up to 1 April 2020, this review lists, not only main, but all targeted therapies in phases II-IV of 257 clinical trials on adults with newly diagnosed or recurrent GBMs for the last twenty years. It does not involve targeted immunotherapies and therapies targeting tumor cell metabolism, that are well documented in other reviews. Without surprise, the most frequently reported drugs are those targeting (i) EGFR (40 clinical trials), and more generally tyrosine kinase receptors (85 clinical trials) and (ii) VEGF/VEGFR (75 clinical trials of which 53 involving bevacizumab). But many other targets and drugs are of interest. They are all listed and thoroughly described, on an one-on-one basis, in four sections related to targeting (i) GBM stem cells and stem cell pathways, (ii) the growth autonomy and migration, (iii) the cell cycle and the escape to cell death, (iv) and angiogenesis.
Collapse
Affiliation(s)
- Elisabete Cruz Da Silva
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Marie-Cécile Mercier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Nelly Etienne-Selloum
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
- Service de Pharmacie, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Monique Dontenwill
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Laurence Choulier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| |
Collapse
|
6
|
Abstract
Metabolic reprogramming is an important characteristics of glioma, the most common form of malignant brain tumor. In this chapter, we aim to discuss some of the recently discovered metabolic alterations in glioma, including the dysregulated TCA cycle, amino acid, nucleotide, and lipid metabolism. We have also detailed some of the metabolomic applications in gliomas, particularly the analyses of body fluids and tissues of glioma patients. With new improvement of the technology, metabolomics will become a powerful tool to discover truly meaningful biomarkers for clinical applications in gliomas. Metabolomic studies of gliomas will also facilitate a better understanding of the molecular targets/pathways and the development of new therapeutic treatments for this devastating disease.
Collapse
|
7
|
Riboni L, Abdel Hadi L, Navone SE, Guarnaccia L, Campanella R, Marfia G. Sphingosine-1-Phosphate in the Tumor Microenvironment: A Signaling Hub Regulating Cancer Hallmarks. Cells 2020; 9:E337. [PMID: 32024090 PMCID: PMC7072483 DOI: 10.3390/cells9020337] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
As a key hub of malignant properties, the cancer microenvironment plays a crucial role intimately connected to tumor properties. Accumulating evidence supports that the lysophospholipid sphingosine-1-phosphate acts as a key signal in the cancer extracellular milieu. In this review, we have a particular focus on glioblastoma, representative of a highly aggressive and deleterious neoplasm in humans. First, we highlight recent advances and emerging concepts for how tumor cells and different recruited normal cells contribute to the sphingosine-1-phosphate enrichment in the cancer microenvironment. Then, we describe and discuss how sphingosine-1-phosphate signaling contributes to favor cancer hallmarks including enhancement of proliferation, stemness, invasion, death resistance, angiogenesis, immune evasion and, possibly, aberrant metabolism. We also discuss the potential of how sphingosine-1-phosphate control mechanisms are coordinated across distinct cancer microenvironments. Further progress in understanding the role of S1P signaling in cancer will depend crucially on increasing knowledge of its participation in the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Loubna Abdel Hadi
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
- Department of Clinical Sciences and Community Health, University of Milan, 20100 Milan, Italy
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| |
Collapse
|
8
|
Qi C, Li Y, Fan X, Jiang Y, Wang R, Yang S, Meng L, Jiang T, Li S. A quantitative SVM approach potentially improves the accuracy of magnetic resonance spectroscopy in the preoperative evaluation of the grades of diffuse gliomas. Neuroimage Clin 2019; 23:101835. [PMID: 31035232 PMCID: PMC6487359 DOI: 10.1016/j.nicl.2019.101835] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 04/13/2019] [Accepted: 04/20/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVES To investigate the association between proton magnetic resonance spectroscopy (1H-MRS) metabolic features and the grade of gliomas, and to establish a machine-learning model to predict the glioma grade. METHODS This study included 112 glioma patients who were divided into the training (n = 74) and validation (n = 38) sets based on the time of hospitalization. Twenty-six metabolic features were extracted from the preoperative 1H-MRS image. The Student's t-test was conducted to screen for differentially expressed features between low- and high-grade gliomas (WHO grades II and III/IV, respectively). Next, the minimum Redundancy Maximum Relevance (mRMR) algorithm was performed to further select features for a support vector machine (SVM) classifier building. Performance of the predictive model was evaluated both in the training and validation sets using ROC curve analysis. RESULTS Among the extracted 1H-MRS metabolic features, thirteen features were differentially expressed. Four features were further selected as grade-predictive imaging signatures using the mRMR algorithm. The predictive performance of the machine-learning model measured by the AUC was 0.825 and 0.820 in the training and validation sets, respectively. This was better than the predictive performances of individual metabolic features, the best of which was 0.812. CONCLUSIONS 1H-MRS metabolic features could help in predicting the grade of gliomas. The machine-learning model achieved a better prediction performance in grading gliomas than individual features, indicating that it could complement the traditionally used metabolic features.
Collapse
Affiliation(s)
- Chong Qi
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Yiming Li
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Xing Fan
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Yin Jiang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Rui Wang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Song Yang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Lanxi Meng
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; National Clinical Research Center for Neurological Diseases, Beijing, China; Center of Brain Tumor, Beijing Institute for Brain Disorders, China; Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), China.
| | - Shaowu Li
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
9
|
Noch EK, Ramakrishna R, Magge R. Challenges in the Treatment of Glioblastoma: Multisystem Mechanisms of Therapeutic Resistance. World Neurosurg 2018; 116:505-517. [PMID: 30049045 DOI: 10.1016/j.wneu.2018.04.022] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/13/2018] [Indexed: 01/14/2023]
Abstract
Glioblastoma is one of the most lethal human cancers, with poor survival despite surgery, radiation treatment, and chemotherapy. Advances in the treatment of this type of brain tumor are limited because of several resistance mechanisms. Such mechanisms involve limited drug entry into the central nervous system compartment by the blood-brain barrier and by actions of the normal brain to counteract tumor-targeting medications. In addition, the vast heterogeneity in glioblastoma contributes to significant therapeutic resistance by preventing adequate control of the entire tumor mass by a single drug and by facilitating escape mechanisms from targeted agents. The stem cell-like characteristics of glioblastoma promote resistance to chemotherapy, radiation, and immunotherapy through upregulation of efflux transporters, promotion of glioblastoma stem cell proliferation in neurogenic zones, and immune suppression, respectively. Metabolic cascades in glioblastoma prevent effective treatments through the optimization of glucose use, the use of alternative nutrient precursors for energy production, and the induction of hypoxia to enhance tumor growth. In the era of precision medicine, an assortment of molecular techniques is being developed to target an individual's unique tumor, with the hope that this personalized strategy will bypass therapeutic resistance. Although each resistance mechanism presents an array of challenges to effective treatment of glioblastoma, as the field recognizes and addresses these difficulties, future treatments may have more efficacy and promise for patients with glioblastoma.
Collapse
Affiliation(s)
- Evan K Noch
- Department of Neurology, Weill Cornell Medical College, New York, New York, USA
| | - Rohan Ramakrishna
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York, USA.
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
10
|
Wang L, Guo S, Zhang H. MiR-98 Promotes Apoptosis of Glioma Cells via Suppressing IKBKE/NF-κB Pathway. Technol Cancer Res Treat 2017; 16:1226-1234. [PMID: 29333957 PMCID: PMC5762096 DOI: 10.1177/1533034617745761] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The inhibitor of kappa B kinase epsilon is overexpressed in glioma and plays antiapoptotic role via activating nuclear factor-kappa B. microRNA-98 can suppress glioma, modulate the activities of nuclear factor-kappa B, and bind to the 3′-untranslated region of inhibitor of kappa B kinase epsilon messenger RNA. This study was aimed to investigate the modulation of inhibitor of kappa B kinase epsilon/nuclear factor-kappa B by microRNA-98 in glioma. The results indicated that microRNA-98 was downregulated in glioma cell lines and human glioma tissues. Overexpression of microRNA-98 in U87MG and T98G glioma cells significantly increased the apoptosis induced by ultraviolet irradiation and suppressed nuclear factor-kappa B luciferase activity, nuclear factor-kappa B p50 subunit expression, and B-cell lymphoma-2 (Bcl-2) expression in glioma cells. Silencing inhibitor of kappa B kinase epsilon decreased the expression of nuclear factor-kappa B p50 subunit and the luciferase activity of nuclear factor-kappa B, while the nuclear factor-kappa B activity could be significantly retrieved when inhibitor of kappa B kinase epsilon was expressed in microRNA-98-transfected cells. These findings indicated that microRNA-98 could promote apoptosis of glioma cells via inhibiting inhibitor of kappa B kinase epsilon/nuclear factor-kappa B signaling and presented a novel regulatory pathway of microRNA-98 by direct suppression of inhibitor of kappa B kinase epsilon/nuclear factor-kappa B expression in glioma cells.
Collapse
Affiliation(s)
- Lingyan Wang
- 1 The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shaolei Guo
- 1 The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Heng Zhang
- 1 The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
11
|
Wang D, Guo M, Yu J, Wang X, Zhang Q, Yang X, Li J, Zhao C, Feng B. Glioma targeting peptide in combination with the P53 C terminus inhibits glioma cell proliferation in vitro. Cytotechnology 2017; 70:153-161. [PMID: 28879517 DOI: 10.1007/s10616-017-0122-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 07/07/2017] [Indexed: 10/18/2022] Open
Abstract
Glioma is a prevalent malignant primary brain tumor in adults, the treatment for which remains a challenge due to its high infiltration and recurrence. Hence, treatments that lead to the suppression of glioma cell migration and invasion may be used in addition to surgery to increase the therapeutic outcome. In this study, we aimed to construct a multifunctional protein that would exert an effect on glioma cell proliferation and migration. The protein is named GL1-P53C-11R and it consists of the glioma-targeting peptide GL1 (G), the P53 C terminus (Pc) and the cell-penetrating peptide arginine (R). GL1-P53C-R was expressed with the fusion protein ZZ and immunofluorescence analysis showed effective delivery of the fused ZZ-GL1-P53C-R protein represented as ZZ-GPcR. The ZZ-GPcR exhibited an inhibitory effect on the proliferation, migration and invasion of U87ΔEGFR cells. Western blotting results indicated that it caused significant changes in the expression levels of cell cycle and apoptotic proteins. Flow cytometric analysis showed increase apoptosis. Our findings suggest that the P53C in the fusion protein ZZ-GPcR can enter into glioma cells to exert its inhibitory effect.
Collapse
Affiliation(s)
- Dan Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Meihua Guo
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Jiawen Yu
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Xinying Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Qian Zhang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Xu Yang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Jiaqi Li
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Chunhui Zhao
- College of Life Sciences, Liaoning Normal University, Dalian, 116029, China.
| | - Bin Feng
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
12
|
Ward NP, Poff AM, Koutnik AP, D’Agostino DP. Complex I inhibition augments dichloroacetate cytotoxicity through enhancing oxidative stress in VM-M3 glioblastoma cells. PLoS One 2017. [PMID: 28644886 PMCID: PMC5482478 DOI: 10.1371/journal.pone.0180061] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The robust glycolytic metabolism of glioblastoma multiforme (GBM) has proven them susceptible to increases in oxidative metabolism induced by the pyruvate mimetic dichloroacetate (DCA). Recent reports demonstrate that the anti-diabetic drug metformin enhances the damaging oxidative stress associated with DCA treatment in cancer cells. We sought to elucidate the role of metformin's reported activity as a mitochondrial complex I inhibitor in the enhancement of DCA cytotoxicity in VM-M3 GBM cells. Metformin potentiated DCA-induced superoxide production, which was required for enhanced cytotoxicity towards VM-M3 cells observed with the combination. Similarly, rotenone enhanced oxidative stress resultant from DCA treatment and this too was required for the noted augmentation of cytotoxicity. Adenosine monophosphate kinase (AMPK) activation was not observed with the concentration of metformin required to enhance DCA activity. Moreover, addition of an activator of AMPK did not enhance DCA cytotoxicity, whereas an inhibitor of AMPK heightened the cytotoxicity of the combination. Our data indicate that metformin enhancement of DCA cytotoxicity is dependent on complex I inhibition. Particularly, that complex I inhibition cooperates with DCA-induction of glucose oxidation to enhance cytotoxic oxidative stress in VM-M3 GBM cells.
Collapse
Affiliation(s)
- Nathan P. Ward
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, United States of America
| | - Angela M. Poff
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, United States of America
| | - Andrew P. Koutnik
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, United States of America
| | - Dominic P. D’Agostino
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL, United States of America
- * E-mail:
| |
Collapse
|
13
|
Strickland M, Stoll EA. Metabolic Reprogramming in Glioma. Front Cell Dev Biol 2017; 5:43. [PMID: 28491867 PMCID: PMC5405080 DOI: 10.3389/fcell.2017.00043] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/07/2017] [Indexed: 12/14/2022] Open
Abstract
Many cancers have long been thought to primarily metabolize glucose for energy production—a phenomenon known as the Warburg Effect, after the classic studies of Otto Warburg in the early twentieth century. Yet cancer cells also utilize other substrates, such as amino acids and fatty acids, to produce raw materials for cellular maintenance and energetic currency to accomplish cellular tasks. The contribution of these substrates is increasingly appreciated in the context of glioma, the most common form of malignant brain tumor. Multiple catabolic pathways are used for energy production within glioma cells, and are linked in many ways to anabolic pathways supporting cellular function. For example: glycolysis both supports energy production and provides carbon skeletons for the synthesis of nucleic acids; meanwhile fatty acids are used both as energetic substrates and as raw materials for lipid membranes. Furthermore, bio-energetic pathways are connected to pro-oncogenic signaling within glioma cells. For example: AMPK signaling links catabolism with cell cycle progression; mTOR signaling contributes to metabolic flexibility and cancer cell survival; the electron transport chain produces ATP and reactive oxygen species (ROS) which act as signaling molecules; Hypoxia Inducible Factors (HIFs) mediate interactions with cells and vasculature within the tumor environment. Mutations in the tumor suppressor p53, and the tricarboxylic acid cycle enzymes Isocitrate Dehydrogenase 1 and 2 have been implicated in oncogenic signaling as well as establishing metabolic phenotypes in genetically-defined subsets of malignant glioma. These pathways critically contribute to tumor biology. The aim of this review is two-fold. Firstly, we present the current state of knowledge regarding the metabolic strategies employed by malignant glioma cells, including aerobic glycolysis; the pentose phosphate pathway; one-carbon metabolism; the tricarboxylic acid cycle, which is central to amino acid metabolism; oxidative phosphorylation; and fatty acid metabolism, which significantly contributes to energy production in glioma cells. Secondly, we highlight processes (including the Randle Effect, AMPK signaling, mTOR activation, etc.) which are understood to link bio-energetic pathways with oncogenic signals, thereby allowing the glioma cell to achieve a pro-malignant state.
Collapse
Affiliation(s)
- Marie Strickland
- Institute of Neuroscience, Newcastle UniversityNewcastle upon Tyne, UK
| | - Elizabeth A Stoll
- Institute of Neuroscience, Newcastle UniversityNewcastle upon Tyne, UK
| |
Collapse
|
14
|
He T, Qiu T, Wang X, Gui H, Wang X, Hu Q, Xia H, Qi G, Wu J, Ma H. Multivoxel magnetic resonance spectroscopy identifies enriched foci of cancer stem-like cells in high-grade gliomas. Onco Targets Ther 2017; 10:195-203. [PMID: 28115854 PMCID: PMC5221654 DOI: 10.2147/ott.s118834] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE This study investigated the correlation between choline/creatine (Cho/Cr) ratios determined by multivoxel proton magnetic resonance spectroscopy (1H-MRS) and the distribution of cancer stem-like cells (CSLCs) in high-grade gliomas. PATIENTS AND METHODS Sixteen patients with high-grade gliomas were recruited and underwent 1H-MRS examination before surgery to identify distinct tumor regions with variable Cho/Cr ratios. Using intraoperative neuronavigation, tumor tissues were accurately sampled from regions with high and low Cho/Cr ratios within each tumor. The distribution of CSLCs in samples from glioma tissue regions with different Cho/Cr ratios was quantified by neurosphere culture, immunohistochemistry, and Western blot. RESULTS The mean neurosphere formation rate in tissues with high Cho/Cr ratios was significantly increased compared with that in low Cho/Cr ratio tissues (13.94±5.94 per 100 cells vs 8.04±3.99 per 100 cells, P<0.001). Immunohistochemistry indicated that tissues with high Cho/Cr ratios had elevated expression of CD133, nestin, and CD15, relative to low Cho/Cr ratio tissue samples (23.6%±3.8% vs 18.3%±3.3%, 25.2%±4.5% vs 19.8%±2.8%, 24.5%±3.8% vs 17.8%±2.2%, respectively; all P<0.001). Western blot demonstrated that relative CD133 and nestin protein expression in high Cho/Cr ratio regions was significantly higher than that in low Cho/Cr ratio tissue samples (0.50±0.17 vs 0.30±0.08, 0.45±0.13 vs 0.27±0.07, respectively; both P<0.001). The protein expression levels of CD133 and nestin were highly correlated with Cho/Cr ratios (r=0.897 and r=0.861, respectively). CONCLUSION Cho/Cr ratios correlate with the distribution of CSLCs in high-grade gliomas, and this may assist in identifying foci enriched with CSLCs and thus improve the management of high-grade gliomas.
Collapse
Affiliation(s)
- Tao He
- Clinical Medicine College, Ningxia Medical University; Department of Neurosurgery, General Hospital of Ningxia Medical University; Ningxia Key Laboratory of Cerebrocranial Diseases, The National Key Laboratory Incubation Base, Yinchuan
| | - Tianming Qiu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai
| | - Xiaodong Wang
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan, People's Republic of China
| | - Hongxing Gui
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School of Rutgers University, Piscataway, NJ, USA
| | - Xilong Wang
- Department of Neurosurgery, General Hospital of Ningxia Medical University
| | - Qikuan Hu
- Ningxia Key Laboratory of Cerebrocranial Diseases, The National Key Laboratory Incubation Base, Yinchuan; Department of Physiology, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Hechun Xia
- Department of Neurosurgery, General Hospital of Ningxia Medical University
| | - Gaoyang Qi
- Clinical Medicine College, Ningxia Medical University; Department of Neurosurgery, General Hospital of Ningxia Medical University
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai
| | - Hui Ma
- Department of Neurosurgery, General Hospital of Ningxia Medical University
| |
Collapse
|
15
|
Woolf EC, Syed N, Scheck AC. Tumor Metabolism, the Ketogenic Diet and β-Hydroxybutyrate: Novel Approaches to Adjuvant Brain Tumor Therapy. Front Mol Neurosci 2016; 9:122. [PMID: 27899882 PMCID: PMC5110522 DOI: 10.3389/fnmol.2016.00122] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/31/2016] [Indexed: 12/18/2022] Open
Abstract
Malignant brain tumors are devastating despite aggressive treatments such as surgical resection, chemotherapy and radiation therapy. The average life expectancy of patients with newly diagnosed glioblastoma is approximately ~18 months. It is clear that increased survival of brain tumor patients requires the design of new therapeutic modalities, especially those that enhance currently available treatments and/or limit tumor growth. One novel therapeutic arena is the metabolic dysregulation that results in an increased need for glucose in tumor cells. This phenomenon suggests that a reduction in tumor growth could be achieved by decreasing glucose availability, which can be accomplished through pharmacological means or through the use of a high-fat, low-carbohydrate ketogenic diet (KD). The KD, as the name implies, also provides increased blood ketones to support the energy needs of normal tissues. Preclinical work from a number of laboratories has shown that the KD does indeed reduce tumor growth in vivo. In addition, the KD has been shown to reduce angiogenesis, inflammation, peri-tumoral edema, migration and invasion. Furthermore, this diet can enhance the activity of radiation and chemotherapy in a mouse model of glioma, thus increasing survival. Additional studies in vitro have indicated that increasing ketones such as β-hydroxybutyrate (βHB) in the absence of glucose reduction can also inhibit cell growth and potentiate the effects of chemotherapy and radiation. Thus, while we are only beginning to understand the pluripotent mechanisms through which the KD affects tumor growth and response to conventional therapies, the emerging data provide strong support for the use of a KD in the treatment of malignant gliomas. This has led to a limited number of clinical trials investigating the use of a KD in patients with primary and recurrent glioma.
Collapse
Affiliation(s)
- Eric C Woolf
- Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical CenterPhoenix, AZ, USA; School of Life Sciences, Arizona State UniversityTempe, AZ, USA
| | - Nelofer Syed
- The John Fulcher Molecular Neuro-Oncology Laboratory, Division of Brain Sciences, Imperial College London London, UK
| | - Adrienne C Scheck
- Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical CenterPhoenix, AZ, USA; School of Life Sciences, Arizona State UniversityTempe, AZ, USA
| |
Collapse
|
16
|
Brucker DP, Maurer GD, Harter PN, Rieger J, Steinbach JP. FOXO3a orchestrates glioma cell responses to starvation conditions and promotes hypoxia-induced cell death. Int J Oncol 2016; 49:2399-2410. [DOI: 10.3892/ijo.2016.3760] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/21/2016] [Indexed: 11/06/2022] Open
|