51
|
The Selective Acetamidine-Based iNOS Inhibitor CM544 Reduces Glioma Cell Proliferation by Enhancing PARP-1 Cleavage In Vitro. Int J Mol Sci 2019; 20:ijms20030495. [PMID: 30678338 PMCID: PMC6387310 DOI: 10.3390/ijms20030495] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 01/31/2023] Open
Abstract
Gliomas are the most aggressive adult primary brain tumors. Expression of inducible Nitric Oxide Synthase has been reported as a hallmark of chemoresistance in gliomas and several studies have reported that inhibition of inducible Nitric Oxide Synthase could be related to a decreased proliferation of glioma cells. The present work was to analyze the molecular effects of the acetamidine derivative compound 39 (formally CM544, N-(3-{[(1-iminioethyl)amino]methyl}benzyl) prolinamide dihydrochloride), a newly synthetized iNOS inhibitor, in a C6 rat glioma cell model. There is evidence of CM544 selective binding to the iNOS, an event that triggers the accumulation of ROS/RNS, the expression of Nrf-2 and the phosphorylation of MAPKs after 3 h of treatment. In the long run, CM544 leads to the dephosphorylation of p38 and to a massive cleavage of PARP-1, confirming the block of C6 rat glioma cell proliferation in the G1/S checkpoint and the occurrence of necrotic cell death.
Collapse
|
52
|
Vanhove C, Goethals I. Magnetic resonance imaging-guided radiation therapy using animal models of glioblastoma. Br J Radiol 2018; 92:20180713. [PMID: 30563357 DOI: 10.1259/bjr.20180713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma is the most aggressive and most common malignant primary brain tumour in adults and has a high mortality and morbidity. Because local tumour control in glioblastoma patients is still elusive in the majority of patients, there is an urgent need for alternative treatment strategies. However, to implement changes to the existing clinical standard of care, research must be conducted to develop alternative treatment strategies. A novel approach in radiotherapy is the introduction of pre-clinical precision image-guided radiation research platforms. The aim of this review is to give a brief overview of the efforts that have been made in the field of radiation research using animal models of glioblastoma. Because MRI has become the reference imaging technique for treatment planning and assessment of therapeutic responses in glioblastoma patients, we will focus in this review on small animal radiotherapy combined with MRI.
Collapse
Affiliation(s)
- Christian Vanhove
- 1 Department of Electronics and Information Systems, Institute Biomedical Technology (IBiTech), Ghent University , Ghent , Belgium
| | - Ingeborg Goethals
- 2 Department of Nuclear Medicine, Ghent University Hospital , Ghent , Belgium
| |
Collapse
|
53
|
Faithful animal modelling of human glioma by using primary initiating cells and its implications for radiosensitization therapy [ARRIVE 1]. Sci Rep 2018; 8:14191. [PMID: 30242200 PMCID: PMC6154973 DOI: 10.1038/s41598-018-32578-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022] Open
Abstract
It has been reported that the ATM kinase inhibitor KU60019 preferentially radiosensitizes orthotopic high grade gliomas (HGG) driven by established U87 and U1242 cell lines bearing specific TP53 mutations. We wished to determine whether those results could be extended to tumors driven by primary glioma initiating cells (GIC) that closely mimic clinical tumors. Orthotopic HGG were developed in immunodeficient non-obese diabetic-severe combined immunodeficient (NOD-SCID) mice by intracranial injection of primary GIC isolated from the adult glioblastoma COMI (acronym of patient’s name) and the pediatric anaplastic astrocytoma 239/12. Similar to the clinical tumors of origin, the orthotopic tumors COMI and 239/12 displayed different growth properties with a voluminous expansive lesion that exerted considerable mass effect on the adjacent structures and an infiltrating, gliomatosis-like growth pattern with limited compressive attitude, respectively. Significant elongations of median animal survival bearing the adult COMI tumor was observed after one KU60019 convection enhanced delivery followed by total 7.5 Gy of ionizing radiation delivered in fifteen 0.5 Gy fractions, as compared to animals treated with vehicle + ionizing radiation (105 vs 89 days; ratio: 0.847; 95% CI of ratio 0.4969 to 1.198; P:0.0417). Similarly, a trend to increased median survival was observed with the radiosensitized pediatric tumor 239/12 (186 vs 167 days; ratio: 0.8978; 95% CI of ratio: 0.5352 to 1.260; P: 0.0891). Our results indicate that radiosensitization by KU60019 is effective towards different orthotopic gliomas that faithfully mimic the clinical tumors and that multiple GIC-based animal models may be essential to develop novel therapeutic protocols for HGG transferable to the clinics.
Collapse
|
54
|
Bolcaen J, Descamps B, Acou M, Deblaere K, den Broecke CV, Boterberg T, Vanhove C, Goethals I. In Vivo DCE-MRI for the Discrimination Between Glioblastoma and Radiation Necrosis in Rats. Mol Imaging Biol 2018; 19:857-866. [PMID: 28303489 DOI: 10.1007/s11307-017-1071-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE In this study, the potential of semiquantitative and quantitative analysis of dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) was investigated to differentiate glioblastoma (GB) from radiation necrosis (RN) in rats. PROCEDURES F98 GB growth was seen on MRI 8-23 days post-inoculation (n = 15). RN lesions developed 6-8 months post-irradiation (n = 10). DCE-MRI was acquired using a fast low-angle shot (FLASH) sequence. Regions of interest (ROIs) encompassed peripheral contrast enhancement in GB (n = 15) and RN (n = 10) as well as central necrosis within these lesions (GB (n = 4), RN (n = 3)). Dynamic contrast-enhanced time series, obtained from the DCE-MRI data, were fitted to determine four function variables (amplitude A, offset from zero C, wash-in rate k, and wash-out rate D) as well as maximal intensity (ImaxF) and time to peak (TTPF). Secondly, maps of semiquantitative and quantitative parameters (extended Tofts model) were created using Olea Sphere (O). Semiquantitative DCE-MRI parameters included wash-inO, wash-outO, area under the curve (AUCO), maximal intensity (ImaxO), and time to peak (TTPO). Quantitative parameters included the rate constant plasma to extravascular-extracellular space (EES) (K trans), the rate constant EES to plasma (K ep), plasma volume (V p), and EES volume (V e). All (semi)quantitative parameters were compared between GB and RN using the Mann-Whitney U test. ROC analysis was performed. RESULTS Wash-in rate (k) and wash-out rate (D) were significantly higher in GB compared to RN using curve fitting (p = 0.016 and p = 0.014). TTPF and TTPO were significantly lower in GB compared to RN (p = 0.001 and p = 0.005, respectively). The highest sensitivity (87 %) and specificity (80 %) were obtained for TTPF by applying a threshold of 581 s. K trans, K ep, and V e were not significantly different between GB and RN. A trend towards higher V p values was found in GB compared to RN, indicating angiogenesis in GB (p = 0.075). CONCLUSIONS Based on our results, in a rat model of GB and RN, wash-in rate, wash-out rate, and the time to peak extracted from DCE-MRI time series data may be useful to discriminate GB from RN.
Collapse
Affiliation(s)
- Julie Bolcaen
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium.
| | - Benedicte Descamps
- iMinds-IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Marjan Acou
- Department of Radiology and Medical Imaging, Ghent University Hospital, Ghent, Belgium
| | - Karel Deblaere
- Department of Radiology and Medical Imaging, Ghent University Hospital, Ghent, Belgium
| | | | - Tom Boterberg
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Christian Vanhove
- iMinds-IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
55
|
Abstract
Electric fields are known to produce biological effects. Depending on specific frequency, they can stimulate healing, directly damage tissues, or produce anti-mitotic activity. Frequencies of 100-300 KHz have been shown to disrupt mitosis and lead to cellular death. Growth of cancer cell lines, both in vitro and in vivo, was shown to be inhibited by application of the electric fields. In the clinical setting, electric fields are available for treatment of brain tumors, specifically glioblastoma (GBM), through a portable device producing so-called tumor treating fields (TTF). Clinical trials conducted in patients with recurrent and newly diagnosed GBM indicated that this novel treatment modality is active and associated with minimal toxicity. This manuscript will review the available evidence supporting the use of TTF in neuro-oncologic practice.
Collapse
|
56
|
Chang H, Zhang J, Miao Z, Ding Y, Xu X, Zhao X, Xu P, Wang Q, Lin Y. Suppression of the Smurf1 Expression Inhibits Tumor Progression in Gliomas. Cell Mol Neurobiol 2018; 38:421-430. [PMID: 28321604 PMCID: PMC11481853 DOI: 10.1007/s10571-017-0485-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/16/2017] [Indexed: 12/13/2022]
Abstract
Glioblastoma, one of the common malignant brain tumors, results in the highly death, but its underlying molecular mechanisms remain unclear. Smurf1, a member of Nedd4 family of HECT-type ligases, has been reported to contribute to tumorigenicity through several important biological pathways. Recently, it was also found to participate in modulate cellular processes, including morphogenesis, autophagy, growth, and cell migration. In this research, we reported the clinical guiding significance of the expression of Smurf1 in human glioma tissues and cell lines. Western blotting analysis discovered that the expression of Smurf1 was increased with WHO grade. Immunohistochemistry levels discovered that high expression of Smurf1 is closely consistent with poor prognosis of glioma. In addition, suppression of Smurf1 can reduce cell invasion and increase the E-cadherin expression, which is a marker of invasion. Our study firstly demonstrated that Smurf1 may promote glioma cell invasion and suppression of the Smurf1 may provide a novel treatment strategy for glioma.
Collapse
Affiliation(s)
- Hao Chang
- Department of Neurosurgery, Wuxi Second Hospital Affiliated to Nanjing Medical University, 68 Zhongshan Road, Wuxi, 214002, Jiangsu, China
| | - Jingning Zhang
- Department of Neurosurgery, Wuxi Second Hospital Affiliated to Nanjing Medical University, 68 Zhongshan Road, Wuxi, 214002, Jiangsu, China
| | - Zengli Miao
- Department of Neurosurgery, Wuxi Second Hospital Affiliated to Nanjing Medical University, 68 Zhongshan Road, Wuxi, 214002, Jiangsu, China
| | - Yasuo Ding
- Department of Neurosurgery, Taizhou People's Hospital Affiliated to Nantong University, 399 Hailing Road, Taizhou, 225300, Jiangsu, China
| | - Xing Xu
- Department of Neurosurgery, Wuxi Second Hospital Affiliated to Nanjing Medical University, 68 Zhongshan Road, Wuxi, 214002, Jiangsu, China
| | - Xudong Zhao
- Department of Neurosurgery, Wuxi Second Hospital Affiliated to Nanjing Medical University, 68 Zhongshan Road, Wuxi, 214002, Jiangsu, China
| | - Peng Xu
- Department of Pathology, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Qing Wang
- Department of Neurosurgery, Wuxi Second Hospital Affiliated to Nanjing Medical University, 68 Zhongshan Road, Wuxi, 214002, Jiangsu, China
| | - Yuchang Lin
- Department of Neurosurgery, Wuxi Second Hospital Affiliated to Nanjing Medical University, 68 Zhongshan Road, Wuxi, 214002, Jiangsu, China.
| |
Collapse
|
57
|
Keu KV, Witney TH, Yaghoubi S, Rosenberg J, Kurien A, Magnusson R, Williams J, Habte F, Wagner JR, Forman S, Brown C, Allen-Auerbach M, Czernin J, Tang W, Jensen MC, Badie B, Gambhir SS. Reporter gene imaging of targeted T cell immunotherapy in recurrent glioma. Sci Transl Med 2018; 9. [PMID: 28100832 DOI: 10.1126/scitranslmed.aag2196] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/14/2016] [Indexed: 12/12/2022]
Abstract
High-grade gliomas are aggressive cancers that often become rapidly fatal. Immunotherapy using CD8+ cytotoxic T lymphocytes (CTLs), engineered to express both herpes simplex virus type 1 thymidine kinase (HSV1-TK) and interleukin-13 (IL-13) zetakine chimeric antigen receptor (CAR), is a treatment strategy with considerable potential. To optimize this and related immunotherapies, it would be helpful to monitor CTL viability and trafficking to glioma cells. We show that noninvasive positron emission tomography (PET) imaging with 9-[4-[18F]fluoro-3-(hydroxymethyl)butyl]guanine ([18F]FHBG) can track HSV1-tk reporter gene expression present in CAR-engineered CTLs. [18F]FHBG imaging was safe and enabled the longitudinal imaging of T cells stably transfected with a PET reporter gene in patients. Further optimization of this imaging approach for monitoring in vivo cell trafficking should greatly benefit various cell-based therapies for cancer.
Collapse
Affiliation(s)
- Khun Visith Keu
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States.,Division of Nuclear Medicine, Hôpital de la Cité-de-la-Santé de Laval, QC, H7M 3L9, Canada
| | - Timothy H Witney
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States.,Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK
| | - Shahriar Yaghoubi
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States
| | - Jarrett Rosenberg
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States
| | - Anita Kurien
- Neurosurgery, City of Hope, Duarte, CA, 91010, United States
| | | | - John Williams
- Molecular & Medical Pharmacology, UCLA, Los Angeles, CA, 90095, United States
| | - Frezghi Habte
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States
| | - Jamie R Wagner
- Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, United States
| | - Stephen Forman
- Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, United States
| | - Christine Brown
- Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, United States
| | | | - Johannes Czernin
- Molecular & Medical Pharmacology, UCLA, Los Angeles, CA, 90095, United States
| | - Winson Tang
- Sangamo BioSciences Inc, Richmond, CA 94804, United States
| | - Michael C Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, 98145, United States
| | - Behnam Badie
- Neurosurgery, City of Hope, Duarte, CA, 91010, United States
| | - Sanjiv S Gambhir
- Department of Radiology, Molecular Imaging Program, Stanford University, Palo Alto, CA, 94305, United States.,Department of Bioengineering, Department of Materials Science & Engineering, Bio-X, Stanford University, Palo Alto, CA, 94305, United States
| |
Collapse
|
58
|
The mechanisms of malic enzyme 2 in the tumorigenesis of human gliomas. Oncotarget 2018; 7:41460-41472. [PMID: 27166188 PMCID: PMC5173072 DOI: 10.18632/oncotarget.9190] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 04/23/2016] [Indexed: 01/10/2023] Open
Abstract
The high level of resistance of glioblastoma multiforme (GBM) to currently used chemotherapies and other conventional therapies, its invasive characteristics and the presence of stem-like cells are the major factors that make the treatment of GBM difficult. Recent studies have demonstrated that the homeostasis of energy metabolism, glycolysis and mitochondrial oxidation of glucose are important for GBM cell growth and chemo-resistance. However, it is not clear which specific gene(s) are involved in the homeostasis of energy metabolism and invasiveness of GBM cells. We performed a preliminary analysis of data obtained from Gene Expression Omnibus profiles and determined that malic enzyme 2 (ME2) expression was positively associated with WHO grade in human primary gliomas. Hence, we evaluated the detailed working mechanisms of ME2 in human GBM cell processes, including proliferation, cell cycle, invasion, migration, ROS, and ATP production. Our data demonstrated that ME2 was involved in GBM growth, invasion and migration. ME2 has two cofactors, NAD+ or NADP+, which are used to produce NADH and NADPH for ATP production and ROS clearance, respectively. If the catalytic activity of ME2 is determined to be critical for its roles in GBM growth, invasion and migration, small molecule inhibitors of ME2 may be valuable drugs for GBM therapy. We hope that our current data provides a candidate treatment strategy for GBM.
Collapse
|
59
|
Liu C, Chen J, Zhu Y, Gong X, Zheng R, Chen N, Chen D, Yan H, Zhang P, Zheng H, Sheng Z, Song L. Highly Sensitive MoS 2-Indocyanine Green Hybrid for Photoacoustic Imaging of Orthotopic Brain Glioma at Deep Site. NANO-MICRO LETTERS 2018; 10:48. [PMID: 30393697 PMCID: PMC6199097 DOI: 10.1007/s40820-018-0202-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/08/2018] [Indexed: 04/14/2023]
Abstract
Photoacoustic technology in combination with molecular imaging is a highly effective method for accurately diagnosing brain glioma. For glioma detection at a deeper site, contrast agents with higher photoacoustic imaging sensitivity are needed. Herein, we report a MoS2-ICG hybrid with indocyanine green (ICG) conjugated to the surface of MoS2 nanosheets. The hybrid significantly enhanced photoacoustic imaging sensitivity compared to MoS2 nanosheets. This conjugation results in remarkably high optical absorbance across a broad near-infrared spectrum, redshifting of the ICG absorption peak and photothermal/photoacoustic conversion efficiency enhancement of ICG. A tumor mass of 3.5 mm beneath the mouse scalp was clearly visualized by using MoS2-ICG as a contrast agent for the in vivo photoacoustic imaging of orthotopic glioma, which is nearly twofold deeper than the tumors imaged in our previous report using MoS2 nanosheet. Thus, combined with its good stability and high biocompatibility, the MoS2-ICG hybrid developed in this study has a great potential for high-efficiency tumor molecular imaging in translational medicine.
Collapse
Affiliation(s)
- Chengbo Liu
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Jingqin Chen
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Ying Zhu
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Xiaojing Gong
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Rongqin Zheng
- Department of Medical Ultrasound, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, People's Republic of China
| | - Ningbo Chen
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Dong Chen
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Huixiang Yan
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Peng Zhang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China.
| | - Liang Song
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
60
|
Bolcaen J, Descamps B, Boterberg T, Vanhove C, Goethals I. PET and MRI Guided Irradiation of a Glioblastoma Rat Model Using a Micro-irradiator. J Vis Exp 2017. [PMID: 29364211 DOI: 10.3791/56601] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
For decades, small animal radiation research was mostly performed using fairly crude experimental setups applying simple single-beam techniques without the ability to target a specific or well-delineated tumor volume. The delivery of radiation was achieved using fixed radiation sources or linear accelerators producing megavoltage (MV) X-rays. These devices are unable to achieve sub-millimeter precision required for small animals. Furthermore, the high doses delivered to healthy surrounding tissue hamper response assessment. To increase the translation between small animal studies and humans, our goal was to mimic the treatment of human glioblastoma in a rat model. To enable a more accurate irradiation in a preclinical setting, recently, precision image-guided small animal radiation research platforms were developed. Similar to human planning systems, treatment planning on these micro-irradiators is based on computed tomography (CT). However, low soft-tissue contrast on CT makes it very challenging to localize targets in certain tissues, such as the brain. Therefore, incorporating magnetic resonance imaging (MRI), which has excellent soft-tissue contrast compared to CT, would enable a more precise delineation of the target for irradiation. In the last decade also biological imaging techniques, such as positron emission tomography (PET) gained interest for radiation therapy treatment guidance. PET enables the visualization of e.g., glucose consumption, amino-acid transport, or hypoxia, present in the tumor. Targeting those highly proliferative or radio-resistant parts of the tumor with a higher dose could give a survival benefit. This hypothesis led to the introduction of the biological tumor volume (BTV), besides the conventional gross target volume (GTV), clinical target volume (CTV), and planned target volume (PTV). At the preclinical imaging lab of Ghent University, a micro-irradiator, a small animal PET, and a 7 T small animal MRI are available. The goal was to incorporate MRI-guided irradiation and PET-guided sub-volume boosting in a glioblastoma rat model.
Collapse
Affiliation(s)
- Julie Bolcaen
- Department of Nuclear Medicine, Ghent University Hospital;
| | - Benedicte Descamps
- IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University
| | - Tom Boterberg
- Department of Radiation Oncology, Ghent University Hospital
| | - Christian Vanhove
- IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University
| | | |
Collapse
|
61
|
Ouyang J, Xu H, Li M, Dai X, Fu F, Zhang X, Lan Q. Paeoniflorin exerts antitumor effects by inactivating S phase kinase-associated protein 2 in glioma cells. Oncol Rep 2017; 39:1052-1062. [PMID: 29286139 PMCID: PMC5802027 DOI: 10.3892/or.2017.6175] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 12/11/2017] [Indexed: 12/18/2022] Open
Abstract
Paeoniflorin (PF), a natural compound isolated from Paeoniae radix, has been shown to exert antitumor effects in various types of human cancers including glioma. However, the mechanism of action is not well understood. S-phase kinase-associated protein (Skp)2 functions as an oncogene in many cancers. In the present study, we investigated whether Skp2 mediates the anti-glioma activity of PF. We found that PF inhibited glioma cell proliferation, migration and invasion, and induced G2/M arrest and apoptosis. Skp2 expression was downregulated in glioma cells treated with PF. PF-induced antitumor effects in glioma cells were abolished by Skp2 overexpression but were enhanced by RNA interference of Skp2. Moreover, PF treatment inhibited U87 cell-derived tumor growth in a xenograft mouse model. These results demonstrate that PF exerts its antitumor effects in part by inhibiting Skp2 expression in glioma cells and could be a promising therapeutic agent for glioma therapy.
Collapse
Affiliation(s)
- Jia Ouyang
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Hui Xu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Ming Li
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xingliang Dai
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Fengqing Fu
- Clinical Immunology Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xueguang Zhang
- Clinical Immunology Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qing Lan
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
62
|
Targeting the T-Lak cell originated protein kinase by OTS964 shrinks the size of power-law coded heterogeneous glioma stem cell populations. Oncotarget 2017; 9:3043-3059. [PMID: 29423027 PMCID: PMC5790444 DOI: 10.18632/oncotarget.23077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/14/2017] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma resists chemoradiotherapy, then, recurs to be a fatal space-occupying lesion. The recurrence is caused by re-growing cell populations such as glioma stem cells (GSCs), suggesting that GSC populations should be targeted. This study addressed whether a novel anti-cancer drug, OTS964, an inhibitor for T-LAK cell originated protein kinase (TOPK), is effective in reducing the size of the heterogeneous GSC populations, a power-law coded heterogeneous GSC populations consisting of glioma sphere (GS) clones, by detailing quantitative growth properties. We found that OTS964 killed GS clones while suppressing the growth of surviving GS clones, thus identifying clone-eliminating and growth-disturbing efficacies of OTS964. The efficacies led to a significant size reduction in GS populations in a dose-dependent manner. The surviving GS clones reconstructed GS populations in the following generations; the recovery of GS populations fits a recurrence after the chemotherapy. The recovering GS clones resisted the clone-eliminating effect of OTS964 in sequential exposure during the growth recovery. However, surprisingly, the resistant properties of the recovered-GS clones had been plastically canceled during self-renewal, and then the GS clones had become re-sensitive to OTS964. Thus, OTS964 targets GSCs to eliminate them or suppress their growth, resulting in shrinkage of the power-law coded GSC populations. We propose a therapy focusing on long-term control in recurrence of glioblastoma via reducing the size of the GSC populations by OTS964.
Collapse
|
63
|
Lieberman F. Glioblastoma update: molecular biology, diagnosis, treatment, response assessment, and translational clinical trials. F1000Res 2017; 6:1892. [PMID: 29263783 PMCID: PMC5658706 DOI: 10.12688/f1000research.11493.1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2017] [Indexed: 12/19/2022] Open
Abstract
This is an exciting time in neuro-oncology. Discoveries elucidating the molecular mechanisms of oncogenesis and the molecular subtypes of glioblastoma multiforme (GBM) have led to new diagnostic and classification schemes with more prognostic power than histology alone. Molecular profiling has become part of the standard neuropathological evaluation of GBM. Chemoradiation followed by adjuvant temozolomide remains the standard therapy for newly diagnosed GBM, but survival remains unsatisfactory. Patients with recurrent GBM continue to have a dismal prognosis, but neuro-oncology centers with active clinical trial programs are seeing a small but increasing cadre of patients with longer survival. Molecularly targeted therapeutics, personalized therapy based on molecular profiling of individual tumors, and immunotherapeutic strategies are all being evaluated and refined in clinical trials. Understanding of the molecular mechanisms of tumor-mediated immunosuppression, and specifically interactions between tumor cells and immune effector cells in the tumor microenvironment, has led to a new generation of immunotherapies, including vaccine and immunomodulatory strategies as well as T-cell-based treatments. Molecularly targeted therapies, chemoradiation, immunotherapies, and anti-angiogenic therapies have created the need to develop more reliable neuroimaging criteria for differentiating the effects of therapy from tumor progression and changes in blood–brain barrier physiology from treatment response. Translational clinical trials for patients with GBM now incorporate quantitative imaging using both magnetic resonance imaging and positron emission tomography techniques. This update presents a summary of the current standards for therapy for newly diagnosed and recurrent GBM and highlights promising translational research.
Collapse
Affiliation(s)
- Frank Lieberman
- Neurooncology Program, UPMC Hillman Cancer Center, UPMC Cancer Pavilion, Pittsburgh, PA, USA
| |
Collapse
|
64
|
Vaglini F, Pardini C, Di Desidero T, Orlandi P, Pasqualetti F, Ottani A, Pacini S, Giuliani D, Guarini S, Bocci G. Melanocortin Receptor-4 and Glioblastoma Cells: Effects of the Selective Antagonist ML00253764 Alone and in Combination with Temozolomide In Vitro and In Vivo. Mol Neurobiol 2017; 55:4984-4997. [DOI: 10.1007/s12035-017-0702-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/31/2017] [Indexed: 12/13/2022]
|
65
|
Nikaki A, Angelidis G, Efthimiadou R, Tsougos I, Valotassiou V, Fountas K, Prasopoulos V, Georgoulias P. 18F-fluorothymidine PET imaging in gliomas: an update. Ann Nucl Med 2017; 31:495-505. [PMID: 28612247 PMCID: PMC5517561 DOI: 10.1007/s12149-017-1183-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/31/2017] [Indexed: 01/18/2023]
Abstract
Brain neoplasms constitute a group of tumors with discrete differentiation grades, and therefore, course of disease and prognosis. Magnetic resonance imaging (MRI) remains the gold standard method for the investigation of central nervous system tumors. However, MRI suffers certain limitations, especially if radiation therapy or chemotherapy has been previously applied. On the other hand, given the development of newer radiopharmaceuticals, positron emission tomography (PET) aims to a better investigation of brain tumors, assisting in the clinical management of the patients. In the present review, the potential contribution of radiolabeled fluorothymidine (FLT) imaging for the evaluation of brain tumors will be discussed. In particular, we will present the role of FLT-PET imaging in the depiction of well and poorly differentiated lesions, the assessment of patient prognosis and treatment response, and the recognition of disease recurrence. Moreover, related semi-quantitative and kinetic parameters will be discussed.
Collapse
Affiliation(s)
- Alexandra Nikaki
- Department of Clinical Physiology, KHSHP, 20 Ahvenistontie Str., 13530, Hämeenlinna, Finland.,Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - George Angelidis
- Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Roxani Efthimiadou
- PET/CT Department, Hygeia Hospital, 4 Erythrou Stavrou Str., 15123, Athens, Greece
| | - Ioannis Tsougos
- Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Varvara Valotassiou
- Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Konstantinos Fountas
- Department of Neurosurgery, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Vasileios Prasopoulos
- PET/CT Department, Hygeia Hospital, 4 Erythrou Stavrou Str., 15123, Athens, Greece.,Department of Nuclear Medicine, Hygeia Hospital, 4 Erythrou Stavrou Str., 15123, Athens, Greece
| | - Panagiotis Georgoulias
- Department of Nuclear Medicine, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece.
| |
Collapse
|
66
|
Lau CS, Mahendraraj K, Chamberlain RS. Oligodendrogliomas in pediatric and adult patients: an outcome-based study from the Surveillance, Epidemiology, and End Result database. Cancer Manag Res 2017; 9:159-166. [PMID: 28496364 PMCID: PMC5422333 DOI: 10.2147/cmar.s117799] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Introduction Oligodendrogliomas (OGs) account for <20% of all intracranial tumors and 25% of gliomas. Despite improvements in imaging techniques allowing for earlier diagnosis, OG is rare among the pediatric population. This study examines a large cohort of OG patients in an effort to define the demographic, clinical, and pathologic factors associated with clinical and survival outcomes. Methods Data on 7,001 OG patients were abstracted from the Surveillance, Epidemiology, and End Result (SEER) database (1973–2013). Pediatric patients were defined as ≤19 years old, and adult patients were defined as age ≥20 years. Results Among 7,001 OG patients, 6.5% were pediatric (mean age 12 ± 6 years), and 93.5% were adult (mean age 46 ± 15 years). Overall, OGs were more common among males, with a male-to-female ratio of 1.28:1. Overall, OGs were more common among Caucasians (76.9%) and also among the African American (10.8% pediatric vs. 4.0% adult) and Hispanic (12.8% pediatric vs. 11.8% adult). OGs occurred most commonly in the temporal lobe of pediatric patients and the frontal lobes of adults. Surgical resection was the primary treatment modality for both pediatric and adult populations (70.6% and 40.5%), followed by combined surgery and radiation (19.7% and 41.2%). Surgical resection was associated with significantly improved survival in both groups. Pediatric patients had a lower overall mortality (19.8% vs. 48.5%) and lower cancer-specific mortality (17.6% vs. 36.8%). Conclusion OGs most often present in Caucasian males in their fifth decade of life with tumors >4 cm in size. Children typically present with tumors in the temporal lobe, while adults present with tumors in the frontal lobe. Surgical resection confers a survival advantage among all patients, especially pediatric patients. Overall survival (OS) and cancer-specific survival are higher in the pediatric population. Further studies on novel treatment techniques, including bevacizumab and immunotherapy approaches, are required.
Collapse
Affiliation(s)
- Christine Sm Lau
- Department of Surgery, Saint Barnabas Medical Center, Livingston, NJ, USA.,Saint George's University School of Medicine, Grenada, West Indies
| | | | - Ronald S Chamberlain
- Department of Surgery, Saint Barnabas Medical Center, Livingston, NJ, USA.,Saint George's University School of Medicine, Grenada, West Indies.,Department of Surgery, New Jersey Medical School, Rutgers University, Newark, NJ.,Department of Surgery, Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| |
Collapse
|
67
|
Qin JB, Liu Z, Zhang H, Shen C, Wang XC, Tan Y, Wang S, Wu XF, Tian J. Grading of Gliomas by Using Radiomic Features on Multiple Magnetic Resonance Imaging (MRI) Sequences. Med Sci Monit 2017; 23:2168-2178. [PMID: 28478462 PMCID: PMC5436423 DOI: 10.12659/msm.901270] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background Gliomas are the most common primary brain neoplasms. Misdiagnosis occurs in glioma grading due to an overlap in conventional MRI manifestations. The aim of the present study was to evaluate the power of radiomic features based on multiple MRI sequences – T2-Weighted-Imaging-FLAIR (FLAIR), T1-Weighted-Imaging-Contrast-Enhanced (T1-CE), and Apparent Diffusion Coefficient (ADC) map – in glioma grading, and to improve the power of glioma grading by combining features. Material/Methods Sixty-six patients with histopathologically proven gliomas underwent T2-FLAIR and T1WI-CE sequence scanning with some patients (n=63) also undergoing DWI scanning. A total of 114 radiomic features were derived with radiomic methods by using in-house software. All radiomic features were compared between high-grade gliomas (HGGs) and low-grade gliomas (LGGs). Features with significant statistical differences were selected for receiver operating characteristic (ROC) curve analysis. The relationships between significantly different radiomic features and glial fibrillary acidic protein (GFAP) expression were evaluated. Results A total of 8 radiomic features from 3 MRI sequences displayed significant differences between LGGs and HGGs. FLAIR GLCM Cluster Shade, T1-CE GLCM Entropy, and ADC GLCM Homogeneity were the best features to use in differentiating LGGs and HGGs in each MRI sequence. The combined feature was best able to differentiate LGGs and HGGs, which improved the accuracy of glioma grading compared to the above features in each MRI sequence. A significant correlation was found between GFAP and T1-CE GLCM Entropy, as well as between GFAP and ADC GLCM Homogeneity. Conclusions The combined radiomic feature had the highest efficacy in distinguishing LGGs from HGGs.
Collapse
Affiliation(s)
- Jiang-Bo Qin
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Zhenyu Liu
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China (mainland)
| | - Hui Zhang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Chen Shen
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China (mainland)
| | - Xiao-Chun Wang
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Yan Tan
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Shuo Wang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China (mainland)
| | - Xiao-Feng Wu
- Department of Radiology, First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China (mainland)
| |
Collapse
|
68
|
Insights into molecular therapy of glioma: current challenges and next generation blueprint. Acta Pharmacol Sin 2017; 38:591-613. [PMID: 28317871 DOI: 10.1038/aps.2016.167] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/21/2016] [Indexed: 12/12/2022]
Abstract
Glioma accounts for the majority of human brain tumors. With prevailing treatment regimens, the patients have poor survival rates. In spite of current development in mainstream glioma therapy, a cure for glioma appears to be out of reach. The infiltrative nature of glioma and acquired resistance substancially restrict the therapeutic options. Better elucidation of the complicated pathobiology of glioma and proteogenomic characterization might eventually open novel avenues for the design of more sophisticated and effective combination regimens. This could be accomplished by individually tailoring progressive neuroimaging techniques, terminating DNA synthesis with prodrug-activating genes, silencing gliomagenesis genes (gene therapy), targeting miRNA oncogenic activity (miRNA-mRNA interaction), combining Hedgehog-Gli/Akt inhibitors with stem cell therapy, employing tumor lysates as antigen sources for efficient depletion of tumor-specific cancer stem cells by cytotoxic T lymphocytes (dendritic cell vaccination), adoptive transfer of chimeric antigen receptor-modified T cells, and combining immune checkpoint inhibitors with conventional therapeutic modalities. Thus, the present review captures the latest trends associated with the molecular mechanisms involved in glial tumorigenesis as well as the limitations of surgery, radiation and chemotherapy. In this article we also critically discuss the next generation molecular therapeutic strategies and their mechanisms for the successful treatment of glioma.
Collapse
|
69
|
Ng WP, Liew BS, Idris Z, Rosman AK. Fluorescence-Guided versus Conventional Surgical Resection of High Grade Glioma: A Single-Centre, 7-Year, Comparative Effectiveness Study. Malays J Med Sci 2017; 24:78-86. [PMID: 28894407 DOI: 10.21315/mjms2017.24.2.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/08/2016] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND High grade gliomas (HGGs) are locally invasive brain tumours that carry a dismal prognosis. Although complete resection increases median survival, the difficulty in reliably demonstrating the tumour border intraoperatively is a norm. The Department of Neurosurgery, Hospital Sungai Buloh is the first public hospital in Malaysia to overcome this problem by adopting fluorescence-guided (FG) surgery using 5-aminolevulinic acid (5-ALA). METHODS A total of 74 patients with histologically proven HGGs treated between January 2008 and December 2014, who fulfilled the inclusion criteria, were enrolled. Kaplan-Meier survival estimates and Cox proportional hazard regression were used. RESULTS Significant longer survival time (months) was observed in the FG group compared with the conventional group (12 months versus 8 months, P < 0.020). Even without adjuvant therapy, HGG patients from FG group survived longer than those from the conventional group (8 months versus 3 months, P = 0.006). No significant differences were seen in postoperative Karnofsky performance scale (KPS) between the groups at 6 weeks and 6 months after surgery compared to pre-operative KPS. Cox proportional hazard regression identified four independent predictors of survival: KPS > 80 (P = 0.010), histology (P < 0.001), surgical method (P < 0.001) and adjuvant therapy (P < 0.001). CONCLUSION This study showed a significant clinical benefit for HGG patients in terms of overall survival using FG surgery as it did not result in worsening of post-operative function outcome when compared with the conventional surgical method. We advocate a further multicentered, randomised controlled trial to support these findings before FG surgery can be implemented as a standard surgical adjunct in local practice for the benefit of HGG patients.
Collapse
Affiliation(s)
- Wei Ping Ng
- Department of Neurosurgery, Hospital Sungai Buloh, Jalan Hospital, 47000 Sungai Buloh, Selangor, Malaysia.,Center for Neuroscience Services and Research, Universiti Sains Malaysia, Jalan Sultanah Zainab 2, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Boon Seng Liew
- Department of Neurosurgery, Hospital Sungai Buloh, Jalan Hospital, 47000 Sungai Buloh, Selangor, Malaysia
| | - Zamzuri Idris
- Center for Neuroscience Services and Research, Universiti Sains Malaysia, Jalan Sultanah Zainab 2, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Azmin Kass Rosman
- Department of Neurosurgery, Hospital Sungai Buloh, Jalan Hospital, 47000 Sungai Buloh, Selangor, Malaysia
| |
Collapse
|
70
|
Bae IS, Kim CH, Kim JM, Cheong JH, Ryu JI, Han MH. Correlation of survivin and B-cell lymphoma 2 expression with pathological malignancy and anti-apoptotic properties of glial cell tumors. Biomed Rep 2017; 6:396-400. [PMID: 28413637 DOI: 10.3892/br.2017.861] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/27/2017] [Indexed: 01/17/2023] Open
Abstract
Apoptosis, whose mechanism remains unclear, is regulated by multiple factors. B-cell lymphoma 2 (Bcl-2) is a well-known anti-apoptotic mediator. Survivin is also a recently recognized novel family inhibitor of apoptosis protein, which inhibits apoptosis via a pathway distinct from Bcl-2 family members. Survivin and Bcl-2 are expressed in various types of human cancer. In the present study, survivin and Bcl-2 expression were characterized in glial cell tumors, and the correlation with pathological malignancy and anti-apoptotic properties were investigated. Fifty-eight patients who had undergone surgical resection for glial cell tumors were evaluated. The pathological types of glial cell tumors were categorized according to the World Health Organization classification. Survivin and Bcl-2 expression levels were investigated by western blot analysis, and apoptosis was detected by DNA fragmentation analysis. The anti-apoptotic rate of glial cell tumors was calculated in tumor samples according to the expression of survivin and Bcl-2 or co-expression. Survivin was characterized in 60.3%, and Bcl-2 was expressed in 43.1% of glioma samples. Co-expression of survivin and Bcl-2 was observed in 25.9% of the tumor specimens. Survivin expression in astrocytic tumors was identified to be significantly associated with the pathological grade (P<0.05); however, Bcl-2 was not (P>0.05). Anti-apoptotic rate of glial cell tumors were detected in 91.4, 92.0 and 100% of patients exhibiting survivin, Bcl-2 or co-expression, respectively. However, the difference in anti-apoptotic frequency between the three groups was not identified to be statistically significant (P>0.05). The present study suggests that survivin expression is correlated with pathological grades of gliomas. In addition, the expression of survivin or Bcl-2 exerts potent anti-apoptotic properties in gliomas. Thus, survivin or Bcl-2 may serve as potential targets for inducing the apoptosis of gliomas.
Collapse
Affiliation(s)
- In-Suk Bae
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri-si, Gyeonggi-do 11923, Republic of Korea
| | - Choong-Hyun Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri-si, Gyeonggi-do 11923, Republic of Korea
| | - Jae-Min Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri-si, Gyeonggi-do 11923, Republic of Korea
| | - Jin-Hwan Cheong
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri-si, Gyeonggi-do 11923, Republic of Korea
| | - Je-Il Ryu
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri-si, Gyeonggi-do 11923, Republic of Korea
| | - Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri-si, Gyeonggi-do 11923, Republic of Korea
| |
Collapse
|
71
|
Rajappa P, Cobb WS, Vartanian E, Huang Y, Daly L, Hoffman C, Zhang J, Shen B, Yanowitch R, Garg K, Cisse B, Haddock S, Huse J, Pisapia DJ, Chan TA, Lyden DC, Bromberg JF, Greenfield JP. Malignant Astrocytic Tumor Progression Potentiated by JAK-mediated Recruitment of Myeloid Cells. Clin Cancer Res 2016; 23:3109-3119. [PMID: 28039266 DOI: 10.1158/1078-0432.ccr-16-1508] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 12/20/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022]
Abstract
Purpose: While the tumor microenvironment has been known to play an integral role in tumor progression, the function of nonresident bone marrow-derived cells (BMDC) remains to be determined in neurologic tumors. Here we identified the contribution of BMDC recruitment in mediating malignant transformation from low- to high-grade gliomas.Experimental Design: We analyzed human blood and tumor samples from patients with low- and high-grade gliomas. A spontaneous platelet-derived growth factor (PDGF) murine glioma model (RCAS) was utilized to recapitulate human disease progression. Levels of CD11b+/GR1+ BMDCs were analyzed at discrete stages of tumor progression. Using bone marrow transplantation, we determined the unique influence of BMDCs in the transition from low- to high-grade glioma. The functional role of these BMDCs was then examined using a JAK 1/2 inhibitor (AZD1480).Results: CD11b+ myeloid cells were significantly increased during tumor progression in peripheral blood and tumors of glioma patients. Increases in CD11b+/GR1+ cells were observed in murine peripheral blood, bone marrow, and tumors during low-grade to high-grade transformation. Transient blockade of CD11b+ cell expansion using a JAK 1/2 Inhibitor (AZD1480) impaired mobilization of these cells and was associated with a reduction in tumor volume, maintenance of a low-grade tumor phenotype, and prolongation in survival.Conclusions: We demonstrate that impaired recruitment of CD11b+ myeloid cells with a JAK1/2 inhibitor inhibits glioma progression in vivo and prolongs survival in a murine glioma model. Clin Cancer Res; 23(12); 3109-19. ©2016 AACR.
Collapse
Affiliation(s)
- Prajwal Rajappa
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - William S Cobb
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Emma Vartanian
- Weill Medical College of Cornell University, New York, New York
| | - Yujie Huang
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Laura Daly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Caitlin Hoffman
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Jane Zhang
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Beiyi Shen
- Weill Medical College of Cornell University, New York, New York
| | - Rachel Yanowitch
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Kunal Garg
- Weill Medical College of Cornell University, New York, New York
| | - Babacar Cisse
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Sara Haddock
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason Huse
- Department of Pathology and, Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - David J Pisapia
- Weill Cornell Medical College, Department of Pathology, Division of Neuropathology, New York, New York
| | - Timothy A Chan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David C Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, Cell and Developmental Biology, Weill Cornell Medical College, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Jacqueline F Bromberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
72
|
Oehlke O, Grosu AL. PET/MRI and brain tumors: focus on radiation oncology treatment planning. Clin Transl Imaging 2016. [DOI: 10.1007/s40336-016-0206-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
73
|
Liu J, Xu J, Li H, Sun C, Yu L, Li Y, Shi C, Zhou X, Bian X, Ping Y, Wen Y, Zhao S, Xu H, Ren L, An T, Wang Q, Yu S. miR-146b-5p functions as a tumor suppressor by targeting TRAF6 and predicts the prognosis of human gliomas. Oncotarget 2016; 6:29129-42. [PMID: 26320176 PMCID: PMC4745716 DOI: 10.18632/oncotarget.4895] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/24/2015] [Indexed: 12/05/2022] Open
Abstract
Down-regulation of miR-146b-5p contributes to tumorigenesis in several human cancers. However, the relevance of miR-146b-5p to prognosis, proliferation and apoptosis in gliomas remains unknown. In the present study, we demonstrated that miR-146b-5p expression was inversely correlated with grades and Ki-67 index in 147 human glioma specimens, but positively correlated with patients’ survival. Furthermore, two distinct subgroups of patients with grade I-IV gliomas with different prognoses were identified according to miR-146b-5p expression in our specimens. Cox regression showed that miR-146b-5p was an independent predictor for patients’ survival. Overexpression of miR-146b-5p dramatically suppressed glioma cell proliferation and induced apoptosis. Mechanistically, we validated TRAF6 as a direct functional target of miR-146b-5p and found that miR-146b-5p overexpression significantly decreased phosphorylated TAK1 and IκBα, the pivotal downstream effectors of TRAF6. Moreover, TRAF6 expression was positively correlated with glioma grades and Ki-67 index but inversely correlated with miR-146b-5p expression and predicted poor prognosis of glioma patients. In glioblastoma cell lines, silencing of TRAF6 could mimic the anti-tumor effect of miR-146b-5p. Our findings identify miR-146b-5p as a tumor suppressor and novel prognostic biomarker of gliomas, and suggest miR-146b-5p and TRAF6 as potential therapeutic candidates for malignant gliomas.
Collapse
Affiliation(s)
- Jing Liu
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Jinling Xu
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Huining Li
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Cuiyun Sun
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Lin Yu
- Department of Biochemistry, Basic Medical College of Tianjin Medical University, Tianjin 300070, China
| | - Yanyan Li
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Cuijuan Shi
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Xuexia Zhou
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yifang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yanjun Wen
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Shujun Zhao
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Laboratory of Hormone and Development, Ministry of Health, Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Hui Xu
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Linlin Ren
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Tongling An
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Qian Wang
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Shizhu Yu
- Department of Neuropathology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Tianjin 300052, China.,Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| |
Collapse
|
74
|
Dunn WD, Aerts HJ, Cooper LA, Holder CA, Hwang SN, Jaffe CC, Brat DJ, Jain R, Flanders AE, Zinn PO, Colen RR, Gutman DA. Assessing the Effects of Software Platforms on Volumetric Segmentation of Glioblastoma. JOURNAL OF NEUROIMAGING IN PSYCHIATRY & NEUROLOGY 2016; 1:64-72. [PMID: 29600296 PMCID: PMC5870135 DOI: 10.17756/jnpn.2016-008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Radiological assessments of biologically relevant regions in glioblastoma have been associated with genotypic characteristics, implying a potential role in personalized medicine. Here, we assess the reproducibility and association with survival of two volumetric segmentation platforms and explore how methodology could impact subsequent interpretation and analysis. METHODS Post-contrast T1- and T2-weighted FLAIR MR images of 67 TCGA patients were segmented into five distinct compartments (necrosis, contrast-enhancement, FLAIR, post contrast abnormal, and total abnormal tumor volumes) by two quantitative image segmentation platforms - 3D Slicer and a method based on Velocity AI and FSL. We investigated the internal consistency of each platform by correlation statistics, association with survival, and concordance with consensus neuroradiologist ratings using ordinal logistic regression. RESULTS We found high correlations between the two platforms for FLAIR, post contrast abnormal, and total abnormal tumor volumes (spearman's r(67) = 0.952, 0.959, and 0.969 respectively). Only modest agreement was observed for necrosis and contrast-enhancement volumes (r(67) = 0.693 and 0.773 respectively), likely arising from differences in manual and automated segmentation methods of these regions by 3D Slicer and Velocity AI/FSL, respectively. Survival analysis based on AUC revealed significant predictive power of both platforms for the following volumes: contrast-enhancement, post contrast abnormal, and total abnormal tumor volumes. Finally, ordinal logistic regression demonstrated correspondence to manual ratings for several features. CONCLUSION Tumor volume measurements from both volumetric platforms produced highly concordant and reproducible estimates across platforms for general features. As automated or semi-automated volumetric measurements replace manual linear or area measurements, it will become increasingly important to keep in mind that measurement differences between segmentation platforms for more detailed features could influence downstream survival or radio genomic analyses.
Collapse
Affiliation(s)
- William D. Dunn
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
| | - Hugo J.W.L. Aerts
- Departments of Radiation Oncology and Radiology, Dana-Farber Cancer
Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,
USA
- Department of Biostatistics & Computational Biology, Dana-Farber
Cancer Institute, Boston, MA, USA
| | - Lee A. Cooper
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
- Department Winship Cancer Institute, Emory University, Atlanta, GA,
USA
- Department Biomedical Engineering, Georgia Institute of
Technology/Emory University, Atlanta, GA, USA
| | - Chad A. Holder
- Department of Radiology and Imaging Sciences, Emory University
School of Medicine, Atlanta, GA, USA
| | - Scott N. Hwang
- Department of Diagnostic Imaging Department, St. Jude
Children’s Research Hospital, Memphis, TN, USA
| | - Carle C. Jaffe
- Department of Radiology, Boston University School of Medicine,
Boston, MA, USA
| | - Daniel J. Brat
- Department of Pathology and Laboratory Medicine, Emory University
School of Medicine, Atlanta, GA, USA
| | - Rajan Jain
- Departments of Radiology and Neurosurgery, NYU School of Medicine,
New York, NY, USA
| | - Adam E. Flanders
- Department of Neuroradiology, Thomas Jefferson University
Hospitals, Philadelphia, PA, USA
| | - Pascal O. Zinn
- Department of Neurosurgery, The University of Texas MD Anderson
Cancer Center, Houston, TX, USA
| | - Rivka R. Colen
- Department of Diagnostic Radiology, The University of Texas MD
Anderson Cancer Center, Houston, TX, USA
| | - David A. Gutman
- Departments of Biomedical Informatics and Neurology, Emory
University School of Medicine, Atlanta, GA, USA
- Department Winship Cancer Institute, Emory University, Atlanta, GA,
USA
| |
Collapse
|
75
|
Hernandez R, Sun H, England CG, Valdovinos HF, Barnhart TE, Yang Y, Cai W. ImmunoPET Imaging of CD146 Expression in Malignant Brain Tumors. Mol Pharm 2016; 13:2563-70. [PMID: 27280694 DOI: 10.1021/acs.molpharmaceut.6b00372] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recently, the overexpression of CD146 and its potential as a therapeutic target in high-grade gliomas, the most lethal type of brain cancer, was uncovered. In this study, we describe the generation of (89)Zr-Df-YY146, a novel (89)Zr-labeled monoclonal antibody (mAb) for the targeting and quantification of CD146 expression in a mouse model of glioblastoma, using noninvasive immunoPET imaging. YY146, a high affinity anti-CD146 mAb, was conjugated to deferoxamine (Df) for labeling with the long-lived positron emitter (89)Zr (t1/2: 78.4 h). In vitro assays, including flow cytometry, immunofluorescence microscopy, and Western blot, were performed with two glioblastoma cell lines, U87MG and U251, to determine their CD146 expression levels. Also, YY146 and Df-YY146's CD146-binding affinities were compared using flow cytometry. In vivo CD146-targeting of (89)Zr-Df-YY146 was evaluated by sequential PET imaging, in athymic nude mice bearing subcutaneously implanted U87MG or U251 tumors. CD146 blocking, ex vivo biodistribution, and histological studies were carried out to confirm (89)Zr-Df-YY146 specificity, as well as the accuracy of PET data. In vitro studies exposed elevated CD146 expression levels in U87MG cells, but negligible levels in U251 cells. Flow cytometry revealed no differences in affinity between YY146 and Df-YY146. (89)Zr labeling of Df-YY146 proceeded with excellent yield (∼80%), radiochemical purity (>95%), and specific activity (∼44 GBq/μmol). Longitudinal PET revealed prominent and persistent (89)Zr-Df-YY146 uptake in mice bearing U87MG tumors that peaked at 14.00 ± 3.28%ID/g (n = 4), 48 h post injection of the tracer. Conversely, uptake was significantly lower in CD146-negative U251 tumors (5.15 ± 0.99%ID/g, at 48 h p.i.; n = 4; P < 0.05). Uptake in U87MG tumors was effectively blocked in a competitive inhibition experiment, corroborating the CD146 specificity of (89)Zr-Df-YY146. Finally, ex vivo biodistribution validated the accuracy of PET data and histological examination successfully correlated tracer uptake with in situ CD146 expression. Prominent, persistent, and specific uptake of (89)Zr-Df-YY146 was observed in brain tumors, demonstrating the potential of this radiotracer for noninvasive PET imaging of CD146 expression. In a future clinical scenario, (89)Zr-Df-YY146 may serve as a tool to guide intervention and assess response to CD146-targeted therapies.
Collapse
Affiliation(s)
- Reinier Hernandez
- Department of Medical Physics, University of Wisconsin , Madison, Wisconsin 53705, United States
| | - Haiyan Sun
- Department of Radiology, University of Wisconsin , Madison, Wisconsin 53705, United States
| | - Christopher G England
- Department of Medical Physics, University of Wisconsin , Madison, Wisconsin 53705, United States
| | - Hector F Valdovinos
- Department of Medical Physics, University of Wisconsin , Madison, Wisconsin 53705, United States
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin , Madison, Wisconsin 53705, United States
| | - Yunan Yang
- Department of Radiology, University of Wisconsin , Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Department of Medical Physics, University of Wisconsin , Madison, Wisconsin 53705, United States.,Department of Radiology, University of Wisconsin , Madison, Wisconsin 53705, United States.,University of Wisconsin Carbone Cancer Center , Madison, Wisconsin 53705, United States
| |
Collapse
|
76
|
Leitzen C, Wilhelm-Buchstab T, Schmeel LC, Garbe S, Greschus S, Müdder T, Oberste-Beulmann S, Simon B, Schild HH, Schüller H. MRI during radiotherapy of glioblastoma : Does MRI allow for prognostic stratification? Strahlenther Onkol 2016; 192:481-8. [PMID: 27259515 DOI: 10.1007/s00066-016-0983-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 04/20/2016] [Indexed: 10/21/2022]
Abstract
AIM To evaluate the role of magnetic resonance imaging (MRI) as a predictor for the clinical course in patients with glioblastoma. PATIENTS AND METHODS In 64 patients with glioblastoma undergoing (chemo)radiotherapy MRI studies were obtained before radiation, after 30 gray (Gy), after 60 Gy and during follow-up. MRI findings were assigned to categories: definite progression, questionable progression, no change. Patients were followed clinically. RESULTS At 30 Gy, 23 of 64 patients (36 %) demonstrated definite (dp; n = 15) or questionable (qp; n = 8) progression; in 41/64 (64 %) no change was found compared with preradiation MRI. After radiotherapy at 60 Gy, 26 of 64 (41 %) patients showed dp (n = 18) or qp (n = 8). In 2 cases with qp at the 30 Gy MRI, progress was unquestionable in the 60 Gy MRI study. In the 64 patients, 5 of the 60 Gy MRIs showed dp/qp after being classified as no change at the 30 Gy MRI, 2 of the 30 Gy MRIs showed qp, while the 60 Gy MRI showed tumour regression and 3 fulfilled the criteria for pseudoprogression during ongoing radiotherapy. The 30 Gy study allowed for prognostic stratification: dp/qp compared to stable patients showed median survival of 10.5 versus 20 months. CONCLUSION MR follow-up after 30 Gy in patients undergoing (chemo)radiotherapy for glioblastoma allows prognostic appraisal. Pseudoprogression has to be taken into account, though rare in our setting. Based on these findings, early discussion of treatment modification is possible.
Collapse
Affiliation(s)
- C Leitzen
- Radiologische Klinik, FE Strahlentherapie, Universitätsklinik Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany.
| | - T Wilhelm-Buchstab
- Radiologische Klinik, FE Strahlentherapie, Universitätsklinik Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - L C Schmeel
- Radiologische Klinik, FE Strahlentherapie, Universitätsklinik Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - S Garbe
- Radiologische Klinik, FE Strahlentherapie, Universitätsklinik Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - S Greschus
- Radiologische Klinik, Universitätsklinik Bonn, Bonn, Germany
| | - T Müdder
- Radiologische Klinik, FE Strahlentherapie, Universitätsklinik Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - S Oberste-Beulmann
- Radiologische Klinik, FE Strahlentherapie, Universitätsklinik Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - B Simon
- Radiologische Klinik, FE Strahlentherapie, Universitätsklinik Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | - H H Schild
- Radiologische Klinik, Universitätsklinik Bonn, Bonn, Germany
| | - H Schüller
- Radiologische Klinik, FE Strahlentherapie, Universitätsklinik Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| |
Collapse
|
77
|
Zheng C, Yang K, Zhang M, Zou M, Bai E, Ma Q, Xu R. Specific protein 1 depletion attenuates glucose uptake and proliferation of human glioma cells by regulating GLUT3 expression. Oncol Lett 2016; 12:125-131. [PMID: 27347112 PMCID: PMC4906678 DOI: 10.3892/ol.2016.4599] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/18/2016] [Indexed: 12/02/2022] Open
Abstract
It has been reported previously that the expression of glucose transporter member 3 (GLUT3) is increased in malignant glioma cells compared with normal glial cells. However, the regulating mechanism that causes this phenomenon remains unknown. The present study investigated the regulating role of transcription factor specific protein 1 (Sp1) in GLUT3 expression in a human glioma cell line. In the present study, Sp1 was identified to directly bind to the GLUT3 5′-untranslated region in human glioma U251 cells. Small interfering RNA- and the Sp1-inhibitor mithramycin A-mediated Sp1 knockdown experiments revealed that Sp1 depletion decreased glucose uptake and inhibited cell growth and invasion of U251 cells by downregulating GLUT3 expression. Therefore Sp1 is an important positive regulator for the expression of GLUT3 in human glioma cells, and may explain the overexpression of GLUT3 in U251 cells. These results suggest that Sp1 may have a role in glioma treatment.
Collapse
Affiliation(s)
- Chuanyi Zheng
- Affiliated Bayi Brain Hospital, General Hospital of Beijing Military Region, Southern Medical University, Beijing 100072, P.R. China; Department of Neurosurgery, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan 570102, P.R. China
| | - Kun Yang
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan 570102, P.R. China
| | - Maoying Zhang
- Affiliated Bayi Brain Hospital, General Hospital of Beijing Military Region, Southern Medical University, Beijing 100072, P.R. China; Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510610, P.R. China
| | - Mingming Zou
- Affiliated Bayi Brain Hospital, General Hospital of Beijing Military Region, Southern Medical University, Beijing 100072, P.R. China
| | - Enqi Bai
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical College, Haikou, Hainan 570102, P.R. China
| | - Quanhong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Ruxiang Xu
- Affiliated Bayi Brain Hospital, General Hospital of Beijing Military Region, Southern Medical University, Beijing 100072, P.R. China
| |
Collapse
|
78
|
Zhou C, Yang Z, Yao Z, Yin B, Pan J, Yu Y, Zhu W, Hua W, Mao Y. Segmentation of peritumoral oedema offers a valuable radiological feature of cerebral metastasis. Br J Radiol 2016; 89:20151054. [PMID: 27119727 DOI: 10.1259/bjr.20151054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Peritumoral oedema (PTO) is commonly observed on MRI in malignant brain tumours including brain metastasis (bMET) and glioblastoma multiforme (GBM). This study aimed to differentiate bMET from GBM by comparing the volume ratio of PTO to tumour lesion (Rvol). METHODS 56 patients with solitary bMET or GBM were enrolled, and MRI was analyzed by a semi-automatic methodology based on MATLAB (Mathworks, Natick, MA). The PTO volume (Voedema) was segmented for quantification using T2 fluid-attenuated inversion-recovery images, while the tumour volume was quantified with enhanced T1 images. The quantitative volume of the tumour, PTO and the ratio of PTO to tumour were interpreted using SPSS(®) (IBM Corp., New York, NY; formerly SPSS Inc., Chicago, IL) by considering different locations and pathologies. RESULTS The tumour volumes of supratentorial GBM, supratentorial bMET (supra-bMET) and infratentorial bMET were 32.22 ± 21.9, 18.45 ± 17.28 and 11.40 ± 5.63 ml, respectively. The corresponding Voedema were 44.08 ± 25.84, 73.20 ± 40.35 and 23.74 ± 7.78 ml, respectively. The Voedema difference between supratentorial and infratentorial lesions is significant (p-value = 0.002). Supra-bMET has a smaller tumour volume (p-value = 0.032), but a larger PTO (p-value = 0.007). The ratio of Voedema to the tumour volume in bMET is statistically higher than that in GBM (p-value = 0.015). The cut-off ratio for identifying bMET from GBM is 3.9, with a specificity and sensitivity of 90.0% and 68.8%, respectively. CONCLUSION Segmentation is an efficient method to quantify irregular PTO. bMET possesses more extensive oedema with smaller tumour volume than does GBM. The Rvol is a valuable index to distinguish bMET from GBM. ADVANCES IN KNOWLEDGE This study presents a new method for the quantitation of PTO to differentiate bMET from GBM.
Collapse
Affiliation(s)
- Chengcheng Zhou
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
| | - Zixiao Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
| | - Zhengwei Yao
- Department of Radiology, Huashan Hospital, Shanghai, China
| | - Bo Yin
- Department of Radiology, Huashan Hospital, Shanghai, China
| | - Jiawei Pan
- Department of Radiology, Huashan Hospital, Shanghai, China
| | - Yang Yu
- Department of Radiology, Huashan Hospital, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Fudan University, Shanghai, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
79
|
Qin LJ, Jia YS, Zhang YB, Wang YH. Cyclooxygenase inhibitor induces the upregulation of connexin-43 expression in C6 glioma cells. Biomed Rep 2016; 4:444-448. [PMID: 27073629 DOI: 10.3892/br.2016.613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/02/2016] [Indexed: 12/20/2022] Open
Abstract
The present study was performed to determine whether aspirin, a cyclooxygenase (COX) inhibitor, has an effect on the expression of connexin 43 (Cx43) in C6 glioma cells. Using an in vitro glioma invasion model, the expression of Cx43 protein in C6 cells was significantly increased following aspirin treatment at a dose of 8 mmol/l for 30, 60 and 120 min via western blot analysis. The peak value of the Cx43 expression was observed in C6 cells after 120 min of aspirin treatment, which was significantly reduced by prostaglandin E2 (PGE2). In addition, aspirin also significantly increased the gap junction intercellular communication (GJIC) activity and reduced glioma invasion, which was induced by PGE2. This led to the conclusion that the aspirin-induced glioma invasion decrease may be associated with the increased expression of Cx43 protein and formation of GJIC.
Collapse
Affiliation(s)
- Li-Juan Qin
- Department of Physiology, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Yong-Sen Jia
- College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Yi-Bing Zhang
- Department of Physiology, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Yin-Huan Wang
- Department of Physiology, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
80
|
Fink AZ, Mogil LB, Lipton ML. Advanced neuroimaging in the clinic: critical appraisal of the evidence base. Br J Radiol 2016; 89:20150753. [PMID: 27074623 DOI: 10.1259/bjr.20150753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The shortage of high-quality systematic reviews in the field of radiology limits evidence-based integration of imaging methods into clinical practice and may perpetuate misconceptions regarding the efficacy and appropriateness of imaging techniques for specific applications. Diffusion tensor imaging for patients with mild traumatic brain injury (DTI-mTBI) and dynamic susceptibility contrast MRI for patients with glioma (DSC-glioma) are applications of quantitative neuroimaging, which similarly detect manifestations of disease where conventional neuroimaging techniques cannot. We performed a critical appraisal of reviews, based on the current evidence-based medicine methodology, addressing the ability of DTI-mTBI and DSC-glioma to (a) detect brain abnormalities and/or (b) predict clinical outcomes. 23 reviews of DTI-mTBI and 26 reviews of DSC-glioma met criteria for inclusion. All reviews addressed detection of brain abnormalities, whereas 12 DTI-mTBI reviews and 22 DSC-glioma reviews addressed prediction of a clinical outcome. All reviews were assessed using a critical appraisal worksheet consisting of 19 yes/no questions. Reviews were graded according to the total number of positive responses and the 2011 Oxford Centre for evidence-based medicine levels of evidence criteria. Reviews addressing DTI-mTBI detection had moderate quality, while those addressing DSC-glioma were of low quality. Reviews addressing prediction of outcomes for both applications were of low quality. Five DTI-mTBI reviews, but only one review of DSC-glioma met criteria for classification as a meta-analysis/systematic/quantitative review.
Collapse
Affiliation(s)
- Adam Z Fink
- 1 The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lisa B Mogil
- 1 The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.,2 SUNY Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Michael L Lipton
- 1 The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.,3 Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA.,4 The Dominick P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.,5 Department of Radiology, Montefiore Medical Center, Bronx, NY, USA.,6 Departments of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
81
|
Wang X, Luo T, Ruan M, Liu P, Wang S, Zhu W. Association of the CCDC26 rs4295627 polymorphism with the risk of glioma: Evidence from 7,290 cases and 11,630 controls. Mol Clin Oncol 2016; 4:878-882. [PMID: 27123300 DOI: 10.3892/mco.2016.813] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 02/22/2016] [Indexed: 01/06/2023] Open
Abstract
Published data on the association between the coiled-coil domain-containing 26 (CCDC26) rs4295627 polymorphism and the risk of glioma have been inconclusive. To further investigate this association, a meta-analysis was performed. By a comprehensive literature search using PubMed and EMBASE databases, a total of 16 case-control studies were identified for inclusion in the meta-analysis. Odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to assess this association. Our results confirmed that the risk with allele G was higher compared with that with allele T for glioma. The results indicated that the allele G of rs4295627 polymorphism in the CCDC26 gene was associated with increased risk of glioma in the homozygote model (GG vs. TT, OR=1.936, 95 %CI: 1.500-2.658, P<0.001), the heterozygote model (GT vs. TT, OR=1.323, 95% CI: 1.241-1.412, P=0.206), the dominant model (GG+GT vs. TT, OR=1.375, 95% CI: 1.256-1.505, P=0.026), the recessive model (GG vs. GT+TT, OR=1.769, 95% CI: 1.302-2.403, P<0.001) and the allele model (G vs. T, OR=1.310, 95% CI: 1.185-1.448, P<0.001). Current evidence suggests that the rs4295627 polymorphism in the CCDC26 gene may contribute to glioma susceptibility. However, further case-control studies are required to confirm our results.
Collapse
Affiliation(s)
- Xiangsheng Wang
- Department of Nephrology, Jining Hospital of Traditional Chinese Medicine, Jining, Shandong 272037, P.R. China
| | - Tong Luo
- Department of Neurology, Jining Hospital of Traditional Chinese Medicine, Jining, Shandong 272037, P.R. China
| | - Mingjun Ruan
- Department of Neurology, Jining Hospital of Traditional Chinese Medicine, Jining, Shandong 272037, P.R. China
| | - Pan Liu
- Department of Stomatology, Laiwu Laicheng People's Hospital, Laiwu, Shandong 271199, P.R. China
| | - Shiying Wang
- Department of Oncology, Laiwu Hospital of Traditional Chinese Medicine, Laiwu, Shandong 271199, P.R. China
| | - Wenhao Zhu
- Department of Neurology, Zibo Hospital of Traditional Chinese Medicine, Zibo, Shandong 255300, P.R. China
| |
Collapse
|
82
|
Aftab Q, Sin WC, Naus CC. Reduction in gap junction intercellular communication promotes glioma migration. Oncotarget 2016; 6:11447-64. [PMID: 25926558 PMCID: PMC4484468 DOI: 10.18632/oncotarget.3407] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 02/19/2015] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma Multiforme (GBM), an aggressive form of adult brain tumor, is difficult to treat due to its invasive nature. One of the molecular changes observed in GBM is a decrease in the expression of the gap junction protein Connexin43 (Cx43); however, how a reduction in Cx43 expression contributes to glioma malignancy is still unclear. In this study we examine whether a decrease in Cx43 protein expression has a role in enhanced cell migration, a key feature associated with increased tumorigenicity. We used a 3D spheroid migration model that mimics the in vivo architecture of tumor cells to quantify migration changes. We found that down-regulation of Cx43 expression in the U118 human glioma cell line increased migration by reducing cell-ECM adhesion, and changed the migration pattern from collective to single cell. In addition gap junction intercellular communication (GJIC) played a more prominent role in mediating migration than the cytoplasmic interactions of the C-terminal tail. Live imaging revealed that reducing Cx43 expression enhanced relative migration by increasing the cell speed and affecting the direction of migration. Taken together our findings reveal an unexplored role of GJIC in facilitating collective migration.
Collapse
Affiliation(s)
- Qurratulain Aftab
- Department of Cellular & Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC
| | - Wun-Chey Sin
- Department of Cellular & Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC
| |
Collapse
|
83
|
Kazda T, Bulik M, Pospisil P, Lakomy R, Smrcka M, Slampa P, Jancalek R. Advanced MRI increases the diagnostic accuracy of recurrent glioblastoma: Single institution thresholds and validation of MR spectroscopy and diffusion weighted MR imaging. NEUROIMAGE-CLINICAL 2016; 11:316-321. [PMID: 27298760 PMCID: PMC4893011 DOI: 10.1016/j.nicl.2016.02.016] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/14/2016] [Accepted: 02/22/2016] [Indexed: 01/08/2023]
Abstract
The accurate identification of glioblastoma progression remains an unmet clinical need. The aim of this prospective single-institutional study is to determine and validate thresholds for the main metabolite concentrations obtained by MR spectroscopy (MRS) and the values of the apparent diffusion coefficient (ADC) to enable distinguishing tumor recurrence from pseudoprogression. Thirty-nine patients after the standard treatment of a glioblastoma underwent advanced imaging by MRS and ADC at the time of suspected recurrence — median time to progression was 6.7 months. The highest significant sensitivity and specificity to call the glioblastoma recurrence was observed for the total choline (tCho) to total N-acetylaspartate (tNAA) concentration ratio with the threshold ≥ 1.3 (sensitivity 100.0% and specificity 94.7%). The ADCmean value higher than 1313 × 10− 6 mm2/s was associated with the pseudoprogression (sensitivity 98.3%, specificity 100.0%). The combination of MRS focused on the tCho/tNAA concentration ratio and the ADCmean value represents imaging methods applicable to early non-invasive differentiation between a glioblastoma recurrence and a pseudoprogression. However, the institutional definition and validation of thresholds for differential diagnostics is needed for the elimination of setup errors before implementation of these multimodal imaging techniques into clinical practice, as well as into clinical trials. For an effective salvage treatment, an accurate diagnosis of GBM recurrence is essential. The standard structural MRI has limited sensitivity and specificity to distinguish GBM progression. GBM recurrence is characterized by the ADCmean value ≤ 1313 × 10− 6 mm2/s and the tCho/tNAA ratio ≥ 1.3. An institutional definition of thresholds is needed, if advanced imaging should be used accurately in clinical practice.
Collapse
Affiliation(s)
- Tomas Kazda
- International Clinical Research Center, St. Anne's University Hospital Brno, 656 91 Brno, Czech Republic; Department of Radiation Oncology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; Department of Radiation Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Martin Bulik
- International Clinical Research Center, St. Anne's University Hospital Brno, 656 91 Brno, Czech Republic; Department of Diagnostic Imaging, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; Department of Diagnostic Imaging, St. Anne's University Hospital Brno, 656 91 Brno, Czech Republic
| | - Petr Pospisil
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; Department of Radiation Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Radek Lakomy
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic; Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Martin Smrcka
- Department of Neurosurgery, University Hospital Brno, Brno 625 00, Czech Republic
| | - Pavel Slampa
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; Department of Radiation Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Radim Jancalek
- Department of Neurosurgery, St. Anne's University Hospital Brno, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic; Department of Neurosurgery, St. Anne's University Hospital Brno, 656 91 Brno, Czech Republic.
| |
Collapse
|
84
|
Positron emission tomography of high-grade gliomas. J Neurooncol 2016; 127:415-25. [PMID: 26897013 DOI: 10.1007/s11060-016-2077-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/15/2016] [Indexed: 10/22/2022]
Abstract
High-grade gliomas [HGG (WHO grades III-IV)] are almost invariably fatal. Imaging of HGG is important for orientating diagnosis, prognosis and treatment planning and is crucial for development of novel, more effective therapies. Given the potentially unlimited number of usable tracing molecules and the elevated number of available radionuclides, PET allows gathering multiple informations on HGG including data on tissue metabolism and drug pharmacokinetics. PET studies on the diagnosis, prognosis and treatment of HGG carried out by most frequently used tracers and radionuclides ((11)C and (18)F) and published in 2014 have been reviewed. These studies demonstrate that a thorough choice of tracers may confer elevated diagnostic and prognostic power to PET imaging of HGG. They also suggest that a combination of PET and MRI may give the most complete and reliable imaging information on HGG and that research on hybrid PET/MRI may be paying back in terms of improved diagnosis, prognosis and treatment planning of these deadly tumours.
Collapse
|
85
|
Yi GZ, Feng WY, Zhou Q, Liu YW, Qi ST. The Impact of MMP-2 and Its Specific Inhibitor TIMP-2 Expression on the WHO Grade and Prognosis of Gliomas in Chinese Population: a Meta-Analysis. Mol Neurobiol 2016; 54:22-30. [PMID: 26729052 PMCID: PMC5219888 DOI: 10.1007/s12035-015-9539-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/09/2015] [Indexed: 01/11/2023]
Abstract
So far, the prognostic value of matrix metalloproteinase 2 (MMP-2) and tissue inhibitor of matrix metalloproteinase 2 (TIMP-2) expressions in patients with gliomas has been widely reported, especially in China. But, the results were inconsistent. Thus, we conducted a meta-analysis to determine the correlation of MMP-2 and TIMP-2 expressions with the prognosis of patients with gliomas. Identical search strategies were used to search relevant literature in electronic databases updated to May 1, 2015, and odds ratios (ORs) with 95 % confidence intervals (95 % CIs) were estimated. Funnel plots and Egger’s tests were conducted for the evaluation of publication bias, and heterogeneity and sensitivity were also analyzed. Finally, a total of 25 studies involving 1572 patients were included in the meta-analysis. Coincidentally, all these studies were conducted in Chinese population. It was found that MMP-2 expression was significantly associated with high-WHO grade gliomas (n = 24, OR = 6.54, CI = 4.98–8.60; I2 = 0 %, P = 0.911) and poor overall survival (OS), while it did not correlate to age (n = 2, OR = 0.78, CI = 0.35–1.74; I2 = 0 %, P = 0.621) and gender (n = 2, OR = 1.15, CI = 0.51–2.62; I2 = 0 %, P = 0.995). Moreover, the results of the pooled analysis indicated that there was no association between TIMP-2 expression and the WHO grade of gliomas (n = 7, OR = 1.02, 95 % CI = 0.68–1.54; I2 = 71.4 %, P = 0.002), but the ratio of MMP-2 and TIMP-2 (MMP-2/TIMP-2) rose with the increase of the WHO grade of gliomas. In conclusion, there was no correlation between TIMP-2 expression and the WHO grade of gliomas, while MMP-2 expression was potently associated with high-WHO grade of gliomas.
Collapse
Affiliation(s)
- Guo-Zhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Avenue North Road No.1838, Guangzhou, 510515, People's Republic of China
| | - Wen-Yan Feng
- The Second College of Clinical Medicine, Southern Medical University, Avenue North Road No.1838, Guangzhou, 510515, People's Republic of China
| | - Qiang Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Avenue North Road No.1838, Guangzhou, 510515, People's Republic of China
| | - Ya-Wei Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Avenue North Road No.1838, Guangzhou, 510515, People's Republic of China
| | - Song-Tao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Avenue North Road No.1838, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
86
|
Popescu AM, Purcaru SO, Alexandru O, Dricu A. New perspectives in glioblastoma antiangiogenic therapy. Contemp Oncol (Pozn) 2015; 20:109-18. [PMID: 27358588 PMCID: PMC4925727 DOI: 10.5114/wo.2015.56122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/15/2015] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GB) is highly vascularised tumour, known to exhibit enhanced infiltrative potential. One of the characteristics of glioblastoma is microvascular proliferation surrounding necrotic areas, as a response to a hypoxic environment, which in turn increases the expression of angiogenic factors and their signalling pathways (RAS/RAF/ERK/MAPK pathway, PI3K/Akt signalling pathway and WTN signalling cascade). Currently, a small number of anti-angiogenic drugs, extending glioblastoma patients survival, are available for clinical use. Most medications are ineffective in clinical therapy of glioblastoma due to acquired malignant cells or intrinsic resistance, angiogenic receptors cross-activation and redundant intracellular signalling, or the inability of the drug to cross the blood-brain barrier and to reach its target in vivo. Researchers have also observed that GB tumours are different in many aspects, even when they derive from the same tissue, which is the reason for personalised therapy. An understanding of the molecular mechanisms regulating glioblastoma angiogenesis and invasion may be important in the future development of curative therapeutic approaches for the treatment of this devastating disease.
Collapse
Affiliation(s)
| | - Stefana Oana Purcaru
- Unit of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova and Clinical Hospital of Neuropsychiatry Craiova, Craiova, Romania
| | - Anica Dricu
- Unit of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
87
|
Qi X, Wan Y, Zhan Q, Yang S, Wang Y, Cai X. Effect of CDKN2A/B rs4977756 polymorphism on glioma risk: a meta-analysis of 16 studies including 24077 participants. Mamm Genome 2015; 27:1-7. [PMID: 26577493 DOI: 10.1007/s00335-015-9612-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/28/2015] [Indexed: 12/11/2022]
Abstract
So far, epidemiological studies have been performed to investigate the association of CDKN2A/B rs4977756 polymorphism and glioma risk. However, the results from different studies remain inconsistent. To clarify these conflicts and to quantitatively evaluate the effect of rs4977756 polymorphism on glioma risk, a meta-analysis was conducted using relevant published clinical studies about rs4977756 polymorphisms and glioma risk. Relevant studies concerning the association between rs4977756 polymorphism and risk of glioma were included in this meta-analysis. Odds ratio (OR) and 95 % confidence interval (CI) were calculated under fixed or random effects models when appropriate. Subgroup analyses were performed by race. This meta-analysis included 13 studies with a total of 8129 cases and 15,858 controls. The pooled results showed that there was an obvious association of CDKN2A/B rs4977756 polymorphism with risk of glioma in all four comparison models (dominant model/AG + GG vs. AA: OR = 1.36, 95 %CI = 1.20-1.54, p < 0.01; heterozygote comparison/AG vs. AA: OR = 1.31, 95 %CI = 1.12-1.53, p < 0.01; homozygote comparison/GG versus AA: OR = 1.49, 95 %CI = 1.36-1.64, p < 0.01; additive model/G vs. A: OR = 1.23, 95 %CI = 1.18-1.28, p < 0.01, respectively). For the subgroup analyses of ethnicities, similar results were observed in Caucasians. However, the association was not found between rs4977756 polymorphism and the risk of glioma in all models for the Asian studies. The CDKN2A/B rs4977756 polymorphism is obvious increase the risk of glioma in Caucasians. Future studies are needed to confirm the results in other ethnic populations.
Collapse
Affiliation(s)
- Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, College of Medical Sciences, Zhejiang University, Hangzhou, 310016, China
| | - Yingfeng Wan
- Department of Neurosurgery, Sir Run Run Shaw Hospital, College of Medical Sciences, Zhejiang University, Hangzhou, 310016, China
| | - Qitao Zhan
- Department of Reproductive Endocrinology, Women's Hospital, College of Medical Sciences, Zhejiang University, Hangzhou, 310006, China
| | - Shuxu Yang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, College of Medical Sciences, Zhejiang University, Hangzhou, 310016, China
| | - Yirong Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, College of Medical Sciences, Zhejiang University, Hangzhou, 310016, China
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medical Sciences, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
88
|
Lei X, Chang L, Ye W, Jiang C, Zhang Z. Raf kinase inhibitor protein (RKIP) inhibits the cell migration and invasion in human glioma cell lines in vitro. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:14214-14220. [PMID: 26823735 PMCID: PMC4713521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/22/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To investigate the effects and the potential mechanisms of RKIP on cell migration, invasion and proliferation in human glioma cell lines in vitro. METHODS The RKIP over-expressing and RKIP knockdown human U87 glioma cells were used to reveal the effects of RKIP on human glioma cells migration, invasion and proliferation. After the recombinant plasmid pcDNA3.0-RKIP or RKIP-shRNA was transfected into the cell lines U87 by the means of liposome assay, the cells migration, invasion and proliferation were detected by wound healing, Transwell and MTT assay. Then, the levels of RKIP, MMP-3, MMP-9 and HMGA2 mRNA transcription were measured by means of RT-qPCR and levels of proteins expressions were determined using Western blot. RESULTS The results of MTT assay suggested that the PKIP have little inhibitive effects on glioma cells proliferation (P>0.05). The present paper showed that the migration distances in the group of RKIP-shRNA were markedly increased compared to the pcDNA3.0-RKIP and control. Similarly, the results showed that the numbers of invasion cells in RKIP-shRNA were remarkably increased than the pcDNA3.0-RKIP group and control group. Western blot and RT-qPCR suggested that over-expressions of RKIP lessened the MMP-2, MMP-9 and HMGA2 expression, however, turning down the RKIP expression showed the inverse effects. CONCLUSION RKIP inhibits the cells migrations and invasions. Meanwhile, RKIP might inhibit the glioma cells through inhibiting MMPs and HMAG2 expression. Therefore, we demonstrated that RKIP is an underlying target for the treatment of glioma.
Collapse
Affiliation(s)
- Xuhui Lei
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Liang Chang
- Department of Neurosurgery, The Tumor Hospital of Harbin Medical UniversityHarbin, China
| | - Wei Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Zhiren Zhang
- Departments of Clinical Pharmacy and Cardiology, The Second Affiliated Hospital of Harbin Medical UniversityHarbin, China
| |
Collapse
|
89
|
Affiliation(s)
- Arthur J L Cooper
- Department of Biochemistry and Molecular Biology, New York Medical College, 15 Dana Road, Valhalla, NY 10595, United States
| |
Collapse
|
90
|
Nie XH, Ou-yang J, Xing Y, Li DY, Dong XY, Liu RE, Xu RX. Paeoniflorin inhibits human glioma cells via STAT3 degradation by the ubiquitin-proteasome pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5611-22. [PMID: 26508835 PMCID: PMC4610779 DOI: 10.2147/dddt.s93912] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We investigated the underlying mechanism for the potent proapoptotic effect of paeoniflorin (PF) on human glioma cells in vitro, focusing on signal transducer and activator of transcription 3 (STAT3) signaling. Significant time- and dose-dependent apoptosis and inhibition of proliferation were observed in PF-treated U87 and U251 glioma cells. Expression of STAT3, its active form phosphorylated STAT3 (p-STAT3), and several downstream molecules, including HIAP, Bcl-2, cyclin D1, and Survivin, were significantly downregulated upon PF treatment. Overexpression of STAT3 induced resistance to PF, suggesting that STAT3 was a critical target of PF. Interestingly, rapid downregulation of STAT3 was consistent with its accelerated degradation, but not with its dephosphorylation or transcriptional modulation. Using specific inhibitors, we demonstrated that the prodegradation effect of PF on STAT3 was mainly through the ubiquitin-proteasome pathway rather than via lysosomal degradation. These findings indicated that PF-induced growth suppression and apoptosis in human glioma cells through the proteasome-dependent degradation of STAT3.
Collapse
Affiliation(s)
- Xiao-hu Nie
- Affiliated Bayi Brain Hospital, Southern Medical University, Beijing, People's Republic of China
| | - Jia Ou-yang
- Nanchang University Medical College, Jiangxi, People's Republic of China
| | - Ying Xing
- Department of Gastroenterology, The 98th Hospital of Nanjing Military Command, Huzhou, Zhejiang, People's Republic of China
| | - Dan-yan Li
- Spleen & Stomach Institution, Guangzhou University of Traditional Chinese Medicine, Guangdong, People's Republic of China
| | - Xing-yu Dong
- Affiliated Bayi Brain Hospital, Southern Medical University, Beijing, People's Republic of China
| | - Ru-en Liu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Ru-xiang Xu
- Bayi Brain Hospital, The Military General Hospital of Beijing PLA, Beijing, People's Republic of China
| |
Collapse
|
91
|
The Diagnostic Ability of Follow-Up Imaging Biomarkers after Treatment of Glioblastoma in the Temozolomide Era: Implications from Proton MR Spectroscopy and Apparent Diffusion Coefficient Mapping. BIOMED RESEARCH INTERNATIONAL 2015; 2015:641023. [PMID: 26448943 PMCID: PMC4584055 DOI: 10.1155/2015/641023] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/25/2015] [Accepted: 04/27/2015] [Indexed: 12/02/2022]
Abstract
Objective. To prospectively determine institutional cut-off values of apparent diffusion coefficients (ADCs) and concentration of tissue metabolites measured by MR spectroscopy (MRS) for early differentiation between glioblastoma (GBM) relapse and treatment-related changes after standard treatment. Materials and Methods. Twenty-four GBM patients who received gross total resection and standard adjuvant therapy underwent MRI examination focusing on the enhancing region suspected of tumor recurrence. ADC maps, concentrations of N-acetylaspartate, choline, creatine, lipids, and lactate, and metabolite ratios were determined. Final diagnosis as determined by biopsy or follow-up imaging was correlated to the results of advanced MRI findings. Results. Eighteen (75%) and 6 (25%) patients developed tumor recurrence and pseudoprogression, respectively. Mean time to radiographic progression from the end of chemoradiotherapy was 5.8 ± 5.6 months. Significant differences in ADC and MRS data were observed between those with progression and pseudoprogression. Recurrence was characterized by N-acetylaspartate ≤ 1.5 mM, choline/N-acetylaspartate ≥ 1.4 (sensitivity 100%, specificity 91.7%), N-acetylaspartate/creatine ≤ 0.7, and ADC ≤ 1300 × 10−6 mm2/s (sensitivity 100%, specificity 100%). Conclusion. Institutional validation of cut-off values obtained from advanced MRI methods is warranted not only for diagnosis of GBM recurrence, but also as enrollment criteria in salvage clinical trials and for reporting of outcomes of initial treatment.
Collapse
|
92
|
Xue L, Wang Y, Yue S, Zhang J. Low MiR-149 expression is associated with unfavorable prognosis and enhanced Akt/mTOR signaling in glioma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:11178-11184. [PMID: 26617839 PMCID: PMC4637654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/20/2015] [Indexed: 06/05/2023]
Abstract
MicroRNAs (miRs) play critical roles in the progression of glioma. Previous in vitro studies have described the anti-tumor role of miR-149 in cancer cells including glioma. In this study, we aimed to investigate whether miR-149 is associated with the prognosis of glioma patients. A total of 163 glioma patients who underwent tumor resection were included in our follow-up study. We found that the miR-149 expression was significantly lower in tumor tissues compared with that in normal tissues (P<0.05). Kaplan-Meier and analysis showed that the miR-149 expression status was significantly associated with the survival duration (logrank test, P<0.001), and multivariate Cox regression revealed that patients with low miR-149 expression were exposed to a 1.825 fold higher death risk (HR=1.825, 95% CI=1.031-3.229, P=0.039) compared with those with high miR-149 expression. Further study showed that Akt/mTOR signaling was hyperactive in low miR-149 expressing tissues. Our study thus demonstrates that miR-149 expression in glioma tissues is critically associated with the prognosis of patients, suggesting its potential clinical significance.
Collapse
Affiliation(s)
- Liang Xue
- Department of Neurosurgery, Tianjin Medical University General Hospital Tianjin 300052, P. R. China
| | - Yi Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital Tianjin 300052, P. R. China
| | - Shuyuan Yue
- Department of Neurosurgery, Tianjin Medical University General Hospital Tianjin 300052, P. R. China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital Tianjin 300052, P. R. China
| |
Collapse
|
93
|
Wang Z, Song Q, Xue J, Zhao Y, Qin S. Ubiquitin-specific protease 28 is overexpressed in human glioblastomas and contributes to glioma tumorigenicity by regulating MYC expression. Exp Biol Med (Maywood) 2015. [PMID: 26209720 DOI: 10.1177/1535370215595468] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The transcription factor MYC, which is dysregulated in the majority of gliomas, is difficult to target directly. Deubiquitinase ubiquitin-specific protease 28 (USP28) stabilizes oncogenic factors, including MYC. However, the contribution of USP28 in tumorigenesis, particularly in glioma, is unknown. Here, we determined the expression of USP28 and assessed its clinical significance in human glioma. We found that USP28 is overexpressed in human glioma but not in normal brain tissue. The level of USP28 protein expression in human glioma tissues was directly correlated with glioma grade. Meanwhile, the level of USP28 protein expression in human glioblastoma tissues was inversely correlated with patient survival. Enforced USP28 expression promotes SW1783 glioma cell proliferation. Moreover, gliomas that arose from USP28-transfected SW1783 cells displayed tumorigenicity in nude mouse model systems. Inhibition of USP28 expression in glioblastoma U373 cells suppressed anchorage-independent growth in vitro and tumorigenicity in vivo. Furthermore, USP28 regulates the expression of MYC protein, which is essential in USP28-induced cell growth in glioma cells. These results showed that USP28 is overexpressed in human glioblastomas and it contributes to glioma tumorigenicity. Therefore, USP28 could be a new target of therapy for human malignant glioma.
Collapse
Affiliation(s)
- Zengwu Wang
- Department of Neurosurgery, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Qimin Song
- Department of Neurosurgery, Linyi People's Hospital, Linyi, 276000, Shandong, China
| | - Jian Xue
- Department of Neurosurgery, Jining No.1 People's Hospital, Jining 272002, Shandong, China
| | - Yumei Zhao
- Department of Neurosurgery, Linyi People's Hospital, Linyi, 276000, Shandong, China
| | - Shiqiang Qin
- Department of Neurosurgery, Weifang People's Hospital, Weifang 261041, Shandong, China
| |
Collapse
|
94
|
Wang D, Hu E, Wu P, Yuan W, Xu S, Sun Z, Shi H, Yuan J, Li G, Zhao S. Genetic variant near TERC influencing the risk of gliomas with older age at diagnosis in a Chinese population. J Neurooncol 2015; 124:57-64. [PMID: 26017031 DOI: 10.1007/s11060-015-1819-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/21/2015] [Indexed: 12/15/2022]
Abstract
A recent genome-wide association study has identified an association between rs1920116 near TERC and high-grade glioma in populations of European ancestry. In order to evaluate the effect of the SNP rs1920116 near TERC in the Chinese population, we examined associations of this candidate SNP with glioma in a sample of 1970 Chinese Han individuals. SNP genotype data were available for 980 Chinese glioma patients and 990 healthy controls. Logistic regression analyses were performed to evaluate the association between rs1920116 and glioma risk adjusted for age, gender and stratified by tumor grade where appropriate. The allele G at TERC rs1920116 are risk factors for gliomas, and its association with glioma risk was consistent across tumor subgroups in the Chinese Han population (OR = 1.18-1.21). In order to assess variation in SNP effect size at different patient ages, glioma cases and controls were divided into 3 age strata, in years: <50, 50-59, and 60+. The results of multiple logistic regression analyses indicate that the SNP has age-specific effects on the risk of developing glioma. Our report confirmed the effects of rs1920116 near TERC on glioma occurring in older peoples in the Chinese Han population for the first time. As TERC is a candidate for inter-individual variation in telomere length, our study supports the hypothesis that telomerase-related mechanisms of telomere maintenance are more associated with gliomas that develop later in life.
Collapse
Affiliation(s)
- Dianhong Wang
- Department of Neurosurgery, The First Affiliated Hospital, Harbin Medical University, No.23 of Youzheng Str., Nangang District, Harbin, 150001, Heilongjiang Province, China,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Gao Y, Li L, Song L. Expression of p16 and Survivin in gliomas and their correlation with cell proliferation. Oncol Lett 2015; 10:301-306. [PMID: 26171019 DOI: 10.3892/ol.2015.3180] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/27/2015] [Indexed: 12/24/2022] Open
Abstract
The survival rate of glioma patients is very low, and a lack of effective diagnostic techniques are available at present. The current study aimed to investigate the expression of p16 and Survivin and their association with proliferation and apoptosis in gliomas, as well as patient characteristics and prognosis. In total, 62 glioma specimens were surgically resected and pathologically confirmed at the Zhumadian Central Hospital (Zhumadian, China) between June 2008 and February 2014. Clinical data, including the gender and age of the patients, as well as the location, infiltration degree, size and pathological stage of the glioma, was collected. In order to evaluate the expression of p16 and Survivin in the gliomas, the Ki-67 labeling index was used to evaluate cell proliferation activity. The number of argyrophilic nucleolar organizer regions and the rate of cellular apoptosis was examined using the terminal deoxynucleotidyl transferase dUTP nick end labeling method. The results were analyzed using SPSS 14.0 statistical software. The positive rate of p16 gene expression in the gliomas was 46.77% (29 cases), and p16 gene expression was positively correlated with the differentiation status, tumor size and pre-operative symptoms. The positive rate of Survivin expression in the gliomas was 69.88% (58 cases), and Survivin expression was positively correlated with tumor size, differentiation status and clinical stage. The proliferation activity of the gliomas was enhanced with increasing p16 and Survivin expression, while apoptosis was inhibited. In conclusion, the overexpression of p16 and Survivin was closely associated with uncontrolled cell proliferation and the inhibition of apoptosis in gliomas. The combined analysis of the expression of p16 and Survivin in gliomas may provide guidance with respect to the clinical diagnosis, evaluation, treatment and prognosis of patients with glioma.
Collapse
Affiliation(s)
- Yansheng Gao
- Department of Neurosurgery Ⅱ, Zhumadian Central Hospital, Zhumadian, Henan 463000, P.R. China
| | - Lingzhen Li
- Department of Neurology Ⅲ, Zhumadian Central Hospital, Zhumadian, Henan 463000, P.R. China
| | - Laijun Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
96
|
Nanoparticle-loaded macrophage-mediated photothermal therapy: potential for glioma treatment. Lasers Med Sci 2015; 30:1357-65. [PMID: 25794592 DOI: 10.1007/s10103-015-1742-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 03/09/2015] [Indexed: 12/13/2022]
Abstract
Gold-based nanoparticles have been used in a number of therapeutic and diagnostic applications. The purpose of this study was to investigate the efficacy of gold-silica nanoshells (AuNS) in photothermal therapy (PTT) of rat gliomas. Rat alveolar macrophages (Ma) were used as nanoparticle delivery vectors. Uptake of AuNS (bare and PEGylated) was investigated in Ma. AuNS were incubated with Ma for 24 h. Phase contrast microscopy was used to visualize the distribution of loaded Ma in three-dimensional glioma spheroids. PTT efficacy was evaluated for both empty (Ma) and AuNS-loaded Ma (Ma(NS)) in both monolayers and spheroids consisting of C6 rat glioma cells and Ma. Monolayers/spheroids were irradiated for 5 min with light from an 810-nm diode laser at irradiances ranging from 7 to 28 W cm(-2). Monolayer survival was evaluated using a 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay while PTT efficacy in spheroids was determined from growth kinetics and live/dead fluorescence microscopy. PTT efficacy was investigated in vivo using a Sprague-Dawley rat glioma model. Five rats received direct intracranial injection of a mixture of 10(4) C6 glioma cells and, 2 days later, an equal number of Ma(NS). Three rats received laser treatment (810 nm; 10 min; 1 W) while the remaining two served as controls (no laser treatment). The uptake ratio of bare to PEGylated AuNS by Ma was 4:1. A significant photothermal effect was observed in vitro, albeit at relatively high radiant exposures (2.1-4.2 kJ cm(-2)). PTT proved effective in vivo in preventing or delaying tumor development in the PTT-treated animals.
Collapse
|
97
|
Lee D, Sun S, Zhang XQ, Zhang PD, Ho ASW, Kiang KMY, Fung CF, Lui WM, Leung GKK. MicroRNA-210 and Endoplasmic Reticulum Chaperones in the Regulation of Chemoresistance in Glioblastoma. J Cancer 2015; 6:227-32. [PMID: 25663939 PMCID: PMC4317757 DOI: 10.7150/jca.10765] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/18/2014] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the commonest primary brain tumour in adults characterized by relentless recurrence due to resistance towards the standard chemotherapeutic agent temozolomide (TMZ). Prolyl 4-hydroxylase, beta polypeptide (P4HB), an endoplasmic reticulum (ER) chaperone, is known to be upregulated in TMZ-resistant GBM cells. MicroRNAs (miRNAs) are non-protein-coding transcripts that may play important roles in GBM chemoresistance. We surmised that miRNA dysregulations may contribute to P4HB upregulation, hence chemoresistance. We found that miRNA-210 (miR-210) was P4HB-targeting and was highly downregulated in TMZ-resistant GBM cells. Forced overexpression of miR-210 led to P4HB downregulation and a reduction in TMZ-resistance. A reciprocal relationship between their expressions was also verified in clinical glioma specimens. Our study is the first to demonstrate a potential link between miR-210 and ER chaperone in determining chemosensitivity in GBM. The findings have important translational implications in suggesting new directions of future studies.
Collapse
Affiliation(s)
- Derek Lee
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Stella Sun
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Xiao Qin Zhang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Ping De Zhang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Amy S W Ho
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Karrie M Y Kiang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Ching Fai Fung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Wai Man Lui
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Gilberto K K Leung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| |
Collapse
|
98
|
Popescu ID, Codrici E, Albulescu L, Mihai S, Enciu AM, Albulescu R, Tanase CP. Potential serum biomarkers for glioblastoma diagnostic assessed by proteomic approaches. Proteome Sci 2014; 12:47. [PMID: 25298751 PMCID: PMC4189552 DOI: 10.1186/s12953-014-0047-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 08/28/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The rapid progress of proteomics over the past years has allowed the discovery of a large number of potential biomarker candidates to improve early tumor diagnosis and therapeutic response, thus being further integrated into clinical environment. High grade gliomas represent one of the most aggressive and treatment-resistant types of human brain cancer, with approximately 9-12 months median survival rate for patients with grade IV glioma (glioblastoma). Using state-of-the-art proteomics technologies, we have investigated the proteome profile for glioblastoma patients in order to identify a novel protein biomarker panel that could discriminate glioblastoma patients from controls and increase diagnostic accuracy. RESULTS In this study, SELDI-ToF MS technology was used to screen potential protein patterns in glioblastoma patients serum; furthermore, LC-MS/MS technology was applied to identify the candidate biomarkers peaks. Through these proteomic approaches, three proteins S100A8, S100A9 and CXCL4 were selected as putative biomarkers and confirmed by ELISA. Next step was to validate the above mentioned molecules as biomarkers through identification of protein expression by Western blot in tumoral versus peritumoral tissue. CONCLUSIONS Proteomic technologies have been used to investigate the protein profile of glioblastoma patients and established several potential diagnostic biomarkers. While it is unlikely for a single biomarker to be highly effective for glioblastoma diagnostic, our data proposed an alternative and efficient approach by using a novel combination of multiple biomarkers.
Collapse
Affiliation(s)
- Ionela Daniela Popescu
- />Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
- />Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, no. 91-95 Splaiul Independentei, 050095 Sector 5, Bucharest, Romania
| | - Elena Codrici
- />Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
| | - Lucian Albulescu
- />Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
- />Current address: Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Simona Mihai
- />Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
| | - Ana-Maria Enciu
- />Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
- />Cellular and Molecular Medicine Department, Carol Davila University of Medicine and Pharmacy, no 8 B-dul Eroilor Sanitari, 050474 Sector 5, Bucharest, Romania
| | - Radu Albulescu
- />Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
- />National Institute for Chemical Pharmaceutical R&D, 112 Calea Vitan, 031299 Sector 3, Bucharest, Romania
| | - Cristiana Pistol Tanase
- />Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
| |
Collapse
|
99
|
The Clinical Utility of Matrix Metalloproteinase 9 in Evaluating Pathological Grade and Prognosis of Glioma Patients: A Meta-Analysis. Mol Neurobiol 2014; 52:38-44. [PMID: 25108671 DOI: 10.1007/s12035-014-8850-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 07/31/2014] [Indexed: 12/14/2022]
Abstract
In the recent years, matrix metalloproteinase 9 (MMP-9) has been focused on as an indicator of glioma grade and prognosis, especially in China. However, all results resulted in many conflicts. So, it is necessary to conduct a meta-analysis to secure a convincing correlation between MMP-9 and grade and prognosis. Eligible studies were included via multiple searches, and then odds ratios (ORs) and hazard ratios (HRs) with 95% confidence intervals (95% CIs) were estimated. Funnel plots were available for evaluation of publication bias. In addition, heterogeneity and sensitivity were also analyzed. In the present meta-analysis, 23 articles were allowed for inclusion with total 1,635 patients. Coincidentally, all studies were conducted in Chinese populations. High MMP-9 expression in gliomas was closely associated with high WHO grade (III+IV) (n = 22, OR = 5.25, 95% CI = 4.09-6.73; p = 0.000), while MMP-9 expression did not correlate to age (n = 4, OR = 1.02, 95 % CI = 0.67-1.54; p = 0.929) and gender (n = 5, OR = 0.91, 95% CI = 0.63-1.33; p = 0.632). Besides, overall survival analysis from two articles revealed MMP-9 expression significantly predicted 5-year-OS (HR = 6.44, 95% CI = 3.88-10.70; p = 0.000) in glioma patients. No heterogeneity and publication bias were observed across all studies. To conclude, this meta-analysis suggests MMP-9 is potently associated with high grade and poor 5 years prognosis, and MMP-9 test of glioma tissues should be established in department of pathology as a routine in clinical practice.
Collapse
|
100
|
Nakamachi T, Sugiyama K, Watanabe J, Imai N, Kagami N, Hori M, Arata S, Shioda S. Comparison of expression and proliferative effect of pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptors on human astrocytoma cell lines. J Mol Neurosci 2014; 54:388-94. [PMID: 25091859 DOI: 10.1007/s12031-014-0362-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 06/24/2014] [Indexed: 12/21/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic neuropeptide considered to be a potent regulator of astrocytes. It has been reported that PACAP also affects astrocytoma cell properties, but the proliferative effects of this peptide in previous reports were inconsistent. The purpose of this study was to search for correlations between malignant potential, PACAP/PACAP receptor expression, and the proliferative potential of four astrocytoma cell lines (KNS-81, KINGS-1, SF-126, and YH-13). Immunohistochemical observations were performed using astrocyte lineage markers with a view to establishing malignant potential, which is inversely correlated to differentiation status in astrocytoma cells. YH-13 showed the most undifferentiated astrocyte-like status, and was immunopositive to a cancer stem cell marker, CD44. These observations suggest that YH-13 is the most malignant of the astrocytoma cell lines tested. Moreover, the strongest PAC1-R immunoreactivity was observed in YH-13 cells. Using real-time PCR analysis, no significant differences among cell lines were detected with respect to PACAP mRNA, but PAC1-R and VPAC1-R mRNA levels were significantly increased in YH-13 cells compared with the other cell lines. Furthermore, when cell lines were treated with PACAP (10(-11) M) for 3 days, the YH-13 cell line, but not of the other cell lines, exhibited a significantly increased cell number. These results suggest that PACAP receptor expression is correlated with the malignant and proliferative potential of astrocytoma cell lines.
Collapse
Affiliation(s)
- Tomoya Nakamachi
- Department of Anatomy, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8555, Japan
| | | | | | | | | | | | | | | |
Collapse
|