1
|
Masiulionytė B, Valiulytė I, Tamašauskas A, Skiriutė D. Metallothionein Genes are Highly Expressed in Malignant Astrocytomas and Associated with Patient Survival. Sci Rep 2019; 9:5406. [PMID: 30932010 PMCID: PMC6443939 DOI: 10.1038/s41598-019-41974-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/18/2019] [Indexed: 02/06/2023] Open
Abstract
Gliomas are heterogeneous, primary brain tumours that originate from glial cells. The main type of gliomas is astrocytomas. There are four grades (I-IV) of astrocytoma malignancy. Astrocytoma grade IV known as glioblastoma multiforme (GBM) is the most common and aggressive type of astrocytic gliomas. Metallothioneins (MT) are low molecular weight, cysteine rich proteins encoded by a family of metallothionein (MT) genes. MT genes play a crucial role in carcinogenesis of diverse malignancies. We proposed MT genes as prognostic markers for malignant astrocytoma. MT1A, MT1E, MT1X, MT2, MT3 gene expression was elevated in grade IV astrocytomas (glioblastomas) as compared to astrocytomas grade I-III. Statistically significant differences were reached for MT1A and MT2 genes (Mann-Whitney test, p < 0.05). High MT1A, MT1X, MT2, MT3 genes expression was associated with shorter patient survival (Log-rank test, p < 0.05). MT1A gene promoter methylation was decreased in glioblastoma (57.6%) while the gene was highly methylated in grade II-III astrocytoma (from 66.7% to 83.3%) and associated with better patient survival (p < 0.05). MT1A gene methylation showed a trend of being associated with higher mRNA expression level in astrocytomas. Increased MT genes expression in grade IV astrocytomas as compared to I-III grade astrocytomas could be associated with malignant tumour behaviour and progression.
Collapse
Affiliation(s)
- Bernadeta Masiulionytė
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, Kaunas, LT-50161, Lithuania
| | - Indrė Valiulytė
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, Kaunas, LT-50161, Lithuania
| | - Arimantas Tamašauskas
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, Kaunas, LT-50161, Lithuania
| | - Daina Skiriutė
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu str. 4, Kaunas, LT-50161, Lithuania.
| |
Collapse
|
2
|
Mazaris P, Hong X, Altshuler D, Schultz L, Poisson LM, Jain R, Mikkelsen T, Rosenblum M, Kalkanis S. Key determinants of short-term and long-term glioblastoma survival: a 14-year retrospective study of patients from the Hermelin Brain Tumor Center at Henry Ford Hospital. Clin Neurol Neurosurg 2014; 120:103-12. [PMID: 24731587 DOI: 10.1016/j.clineuro.2014.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 02/06/2014] [Accepted: 03/01/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Glioblastoma (GBM) is a heterogeneous neoplasm with a small percentage of long-term survivors. Despite aggressive surgical resection and advances in radiotherapy and chemotherapy, the median survival for patients with GBM is 12-14 months. Factors associated with a favorable prognosis include young age, high performance status, gross resection >98%, non-eloquent tumor location and O6-methylguanine methyltransferase (MGMT) promoter methylation. We retrospectively analyzed the relationship of clinical, epidemiologic, genetic and molecular characteristics with survival in patients with GBM. METHODS This retrospective analysis of overall survival looked at the outcomes of 480 patients diagnosed with GBM over 14 years at a single institution. Multivariate analysis was performed examining multiple patient characteristics. RESULTS Median survival time improved from 11.8 months in patients diagnosed from 1995 to 1999 to 15.9 months in those diagnosed from 2005 to 2008. Factors associated with survivor groups were age, KPS, tumor resection, treatment received and early progression. 18 cancer-related genes were upregulated in short-term survivors and five genes were downregulated in short-term survivors. CONCLUSIONS Epidemiologic, clinical, and molecular characteristics all contribute to GBM prognosis. Identifying factors associated with survival is important for treatment strategies as well as research for novel therapeutics and technologies. This study demonstrated improved survival for patients over time as well as significant differences among survivor groups.
Collapse
Affiliation(s)
- Paul Mazaris
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Xin Hong
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - David Altshuler
- Wayne State University School of Medicine, 1313 Scott Hall, Detroit 48201, USA
| | - Lonni Schultz
- Public Health Sciences, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Laila M Poisson
- Public Health Sciences, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Rajan Jain
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA; Radiology, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Tom Mikkelsen
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Mark Rosenblum
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA
| | - Steven Kalkanis
- Departments of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 West Grand Boulevard, Detroit 48202, USA.
| |
Collapse
|
3
|
Liu C, Zhang H, Pan Y, Huang F, Xia S. Towards MIB-1 and p53 detection in glioma magnetic resonance image: a novel computational image analysis method. Phys Med Biol 2012. [PMID: 23202049 DOI: 10.1088/0031-9155/57/24/8393] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Glioma is the primary tumor in the central nervous system, and poses one of the greatest challenges in clinical treatment. MIB-1 and p53 are the most useful biomarkers for gliomas and could help neurosurgeons establish a therapeutic schedule. However, these biomarkers are commonly detected with the help of immunohistochemistry (IHC), which wastes time and energy and is often influenced by subjective factors. To reduce the subjective factors and improve the efficiency in the judgment of IHC, a novel magnetic resonance image (MRI) analysis method is proposed in the present study to detect the expression status of MIB-1 and p53 in IHC. The proposed method includes two kinds of MRI acquisition (FLAIR and T1 FLAIR images), regions of interest (ROIs) selection, texture features (i.e. the gray level gradient co-occurrence matrix (GLGCM), Minkowski functions (MFs), etc) extraction in ROIs, and classification with a support vector machine in a leave-one-out cross validation strategy. By classifying the ROIs, the performance of the method was evaluated by accuracy, area under ROC curve (AUC), etc. A high accuracy (0.7640 ± 0.0225) and AUC (0.7873 ± 0.0377) for MIB-I detection were achieved. In terms of the texture features, 0.7621 ± 0.0199, 0.7666 ± 0.0365 and 0.7426 ± 0.0451 AUC can be obtained using only GLCM, RLM or GLGCM for MIB-1 detection, respectively. In all, the experimental results demonstrated that MR image texture features are associated with the expression status of MIB-1 and p53. The proposed method has the potential to realize high accuracy and robust detection for MIB-I expression status, which makes it promising for clinical glioma diagnosis and prognosis.
Collapse
Affiliation(s)
- Chenbin Liu
- Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | | | | | | | | |
Collapse
|
4
|
Yan H, Yang K, Xiao H, Zou YJ, Zhang WB, Liu HY. Over-expression of cofilin-1 and phosphoglycerate kinase 1 in astrocytomas involved in pathogenesis of radioresistance. CNS Neurosci Ther 2012; 18:729-36. [PMID: 22742733 DOI: 10.1111/j.1755-5949.2012.00353.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/14/2012] [Accepted: 04/19/2012] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Astrocytoma is among the most common intracranial tumors and radiotherapy is typically used after its resection. One of the outstanding problems encountered in the treatment is radioresistance. The lack of efficient biomarkers for evaluating the radiosensitivity of glioma precludes advances in treatment of astrocytoma and remains the most fatal cancer. METHODS To identify potential biomarkers for assessing the radioresistance of astrocytomas, the following study investigated the proteome of astrocytoma in surgical samples from 15 typical patients. The patients were divided into 2 groups: radioresistant vs. radiosensitive (controls). Proteome was assessed using two-dimensional liquid chromatography tandem mass spectrometry (2D-LC-MS/MS). Western blot was adopted to confirm the differential expression of proteins. RESULTS A total of 36 proteins were expressed differently between the 2 groups, represented by cofilin-1 and phosphoglycerate kinase 1 (PGK1), which up-regulated significantly in radioresistant astrocytomas though there was no obvious morphological change of tumors. Western blot analysis revealed elevated levels of protein extracts in radioresistant astrocytomas compared with the radiosensitive group. CONCLUSIONS The results indicated cofilin-1 enhances the motility of tumor cells which is important invasive potential of malignancy. PGK1 is metabolic enzyme and seems to be correlated with the negative prognosis following radiotherapy. Thus, cofilin-1 and PGK1 might be involved in the radioresistant phenotype and are potential biomarkers for developing better therapeutic methods.
Collapse
Affiliation(s)
- Hua Yan
- Department of Neurosurgery, Brain Hospital Affiliated to Nanjing Medical University, China
| | | | | | | | | | | |
Collapse
|
5
|
Locatelli E, Broggi F, Ponti J, Marmorato P, Franchini F, Lena S, Franchini MC. Lipophilic silver nanoparticles and their polymeric entrapment into targeted-PEG-based micelles for the treatment of glioblastoma. Adv Healthc Mater 2012. [PMID: 23184752 DOI: 10.1002/adhm.201100047] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A simple method for the synthesis of lipophilic Ag NPs have been developed. The coated Ag NPs have been entrapped into a FDA-approved and targetable PEG-based polymeric nanoparticles, and this nanocarrier has been conjugated with the peptide chlorotoxin. Uptake experiments have shown a cell-specific recognition of the Ag-1-PNPs-Cltx on U87MG cell lines in comparison to Balb/3T3. The uptake of Ag into the cells was quantified and an interesting cytotoxic effect (IC50 = 45 μM) has been found on glioblastoma cell lines.
Collapse
Affiliation(s)
- Erica Locatelli
- Dipartimento di Chimica Organica, "A. Mangini", University, Viale Risorgimento 4, 40136 Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
6
|
Combined gene expression and protein interaction analysis of dynamic modularity in glioma prognosis. J Neurooncol 2011; 107:281-8. [DOI: 10.1007/s11060-011-0757-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 10/22/2011] [Indexed: 01/27/2023]
|
7
|
Jijiwa M, Demir H, Gupta S, Leung C, Joshi K, Orozco N, Huang T, Yildiz VO, Shibahara I, de Jesus JA, Yong WH, Mischel PS, Fernandez S, Kornblum HI, Nakano I. CD44v6 regulates growth of brain tumor stem cells partially through the AKT-mediated pathway. PLoS One 2011; 6:e24217. [PMID: 21915300 PMCID: PMC3167830 DOI: 10.1371/journal.pone.0024217] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 08/08/2011] [Indexed: 11/24/2022] Open
Abstract
Identification of stem cell-like brain tumor cells (brain tumor stem-like cells; BTSC) has gained substantial attention by scientists and physicians. However, the mechanism of tumor initiation and proliferation is still poorly understood. CD44 is a cell surface protein linked to tumorigenesis in various cancers. In particular, one of its variant isoforms, CD44v6, is associated with several cancer types. To date its expression and function in BTSC is yet to be identified. Here, we demonstrate the presence and function of the variant form 6 of CD44 (CD44v6) in BTSC of a subset of glioblastoma multiforme (GBM). Patients with CD44high GBM exhibited significantly poorer prognoses. Among various variant forms, CD44v6 was the only isoform that was detected in BTSC and its knockdown inhibited in vitro growth of BTSC from CD44high GBM but not from CD44low GBM. In contrast, this siRNA-mediated growth inhibition was not apparent in the matched GBM sample that does not possess stem-like properties. Stimulation with a CD44v6 ligand, osteopontin (OPN), increased expression of phosphorylated AKT in CD44high GBM, but not in CD44low GBM. Lastly, in a mouse spontaneous intracranial tumor model, CD44v6 was abundantly expressed by tumor precursors, in contrast to no detectable CD44v6 expression in normal neural precursors. Furthermore, overexpression of mouse CD44v6 or OPN, but not its dominant negative form, resulted in enhanced growth of the mouse tumor stem-like cells in vitro. Collectively, these data indicate that a subset of GBM expresses high CD44 in BTSC, and its growth may depend on CD44v6/AKTpathway.
Collapse
Affiliation(s)
- Mayumi Jijiwa
- Department of Neurological Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | - Habibe Demir
- Department of Neurological Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | - Snehalata Gupta
- Department of Neurological Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | - Crystal Leung
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kaushal Joshi
- Department of Neurological Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | - Nicholas Orozco
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tiffany Huang
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Vedat O. Yildiz
- Center for Biostatistics, The Ohio State University, Columbus, Ohio, United States of America
| | - Ichiyo Shibahara
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jason A. de Jesus
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - William H. Yong
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Paul S. Mischel
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Soledad Fernandez
- Center for Biostatistics, The Ohio State University, Columbus, Ohio, United States of America
| | - Harley I. Kornblum
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Ichiro Nakano
- Department of Neurological Surgery, The Ohio State University, Columbus, Ohio, United States of America
- James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
8
|
Castro MG, Candolfi M, Kroeger K, King GD, Curtin JF, Yagiz K, Mineharu Y, Assi H, Wibowo M, Ghulam Muhammad AKM, Foulad D, Puntel M, Lowenstein PR. Gene therapy and targeted toxins for glioma. Curr Gene Ther 2011; 11:155-80. [PMID: 21453286 DOI: 10.2174/156652311795684722] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/08/2011] [Indexed: 12/12/2022]
Abstract
The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.
Collapse
Affiliation(s)
- Maria G Castro
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Navaratne KS, Weil RJ. The Aurora A F31I polymorphism is not a risk factor for glioblastoma. World Neurosurg 2011; 74:144-6. [PMID: 21300005 DOI: 10.1016/j.wneu.2010.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 04/05/2010] [Indexed: 10/18/2022]
Abstract
BACKGROUND The Aurora A F31I polymorphism has been linked to increased risk of several human cancers. Glioblastoma (GBM) is the most malignant glioma as well as the most frequent adult primary brain tumor. METHODS In this case-control study we investigated the association of Aurora A F31I polymorphism with GBM risk. RESULTS A total of 96 histologically confirmed patients with GBM and 93 health individuals were analyzed for the Aurora A F31I single nucleotide polymorphism. The single nucleotide polymorphism data were compared and risks associated with individual genotypes and allelotypes were evaluated in terms of odd ratios. CONCLUSIONS The data obtained from this case-control study demonstrates that the Aurora A F31I polymorphism is not a significant risk for GBMs.
Collapse
Affiliation(s)
- Kapila S Navaratne
- The Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery and the Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| | | |
Collapse
|
10
|
Hahm SW, Park J, Son YS. Opuntia humifusa partitioned extracts inhibit the growth of U87MG human glioblastoma cells. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2010; 65:247-252. [PMID: 20814744 DOI: 10.1007/s11130-010-0188-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Opuntia humifusa, a member of the Cactaceae family widely distributed in the southern regions of the Korean peninsula, has potential bioactive functions and medicinal benefits. In the present study, we investigated the effect of hexane, ethyl acetate extracts and water partitioned fraction of O. humifusa on proliferation, G1 arrest and apoptosis in U87MG human glioblastoma cells. Glioblastoma cellular proliferation was evaluated using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, and the effects of O. humifusa partitioned extracts on cell cycle and apoptosis were analyzed by flow cytometry. Our results revealed that when U87MG cells were treated with hexane extracts and water partitioned fraction of O. humifusa, the number of viable cells decreased in a concentration-dependent manner. In addition, water partitioned fractions of O. humifusa induced G1 arrest and non-apoptotic cell death as well as significant increases in ROS production in U87MG cells. In conclusion, water partitioned fractions of O. humifusa induce G1 arrest and inhibit U87MG human glioblastoma cell proliferation.
Collapse
Affiliation(s)
- Sahng-Wook Hahm
- Department of Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Anam-dong, Seongbuk-gu, Seoul 136-713, South Korea
| | | | | |
Collapse
|
11
|
Ilie MI, Hofman V, Ortholan C, Ammadi RE, Bonnetaud C, Havet K, Venissac N, Mouroux J, Mazure NM, Pouysségur J, Hofman P. Overexpression of carbonic anhydrase XII in tissues from resectable non-small cell lung cancers is a biomarker of good prognosis. Int J Cancer 2010; 128:1614-23. [PMID: 20521252 DOI: 10.1002/ijc.25491] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 05/27/2010] [Indexed: 01/14/2023]
Abstract
The pattern of protein expression in tumors is under the influence of nutrient stress, hypoxia and low pH, which determines the survival of neoplastic cells and the development of tumors. Carbonic anhydrase XII (CAXII) is a transmembrane enzyme that catalyzes the reversible hydration of cell-generated carbon dioxide into protons and bicarbonate. Hypoxic conditions activate its transcription and translation and enhanced expression is often present in several types of tumors. The aim of our study was to assess the prognostic significance of CAXII tumor tissues expression in patients with NSCLC. Five hundred fifty-five tumors were immunostained for CAXII on tissue microarrays (TMA) and the results were correlated with clinicopathological parameters and outcome of patients. CAXII overexpression was present in 105/555 (19%) cases and was associated with tumors of lower grade (p = 0.015) and histological type (p < 0.001), being significantly higher in squamous cell carcinoma. High CAXII expression correlated with better overall and disease-specific survival of patients with resectable NSCLC in univariate (p < 0.001) and multivariate survival analyses (p < 0.001). In conclusion, this is the first study demonstrating that a high CAXII tumor tissue expression evaluated on TMAs is related to a better outcome in a large series of patients with resectable NSCLC.
Collapse
Affiliation(s)
- Marius I Ilie
- Laboratory of Clinical and Experimental Pathology, Louis Pasteur Hospital, Nice, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Comes Franchini M, Bonini BF, Camaggi CM, Gentili D, Pession A, Rani M, Strocchi E. Design and synthesis of novel 3,4-disubstituted pyrazoles for nanomedicine applications against malignant gliomas. Eur J Med Chem 2010; 45:2024-33. [DOI: 10.1016/j.ejmech.2010.01.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 01/08/2010] [Accepted: 01/11/2010] [Indexed: 11/28/2022]
|
13
|
Abstract
Glioblastoma multiforme is the most common primary central nervous system tumor. The prognosis for these malignant brain tumors is poor, with a median survival of 14 months and a 5-year survival rate below 2%. Development of novel treatments is essential to improving survival and quality of life for these patients. Endogenous heat shock proteins have been implicated in mediation of both adaptive and innate immunity, and there is a rising interest in the use of this safe and multifaceted heat shock protein vaccine therapy as a promising treatment for human cancers, including glioblastoma multiforme.
Collapse
Affiliation(s)
- Isaac Yang
- Department of Neurological Surgery, University of California at San Francisco, 505 Parnassus Avenue, Room M779, Campus 0112, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
14
|
Sethi D, Kumar A, Gupta KC, Kumar P. A facile method for the construction of oligonucleotide microarrays. Bioconjug Chem 2009; 19:2136-43. [PMID: 18939860 DOI: 10.1021/bc800241k] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In recent years, the oligonucleotide-based microarray technique has emerged as a powerful and promising tool for various molecular biological studies. Here, a facile protocol for the construction of an oligonucleotide microarray is demonstrated that involves immobilization of oligonucleotide-trimethoxysilyl conjugates onto virgin glass microslides. The projected immobilization strategy reflects high immobilization efficiency ( approximately 36-40%) and signal-to-noise ratio ( approximately 98), and hybridization efficiency ( approximately 32-35%). Using the proposed protocol, aminoalkyl, mercaptoalkyl, and phosphorylated oligonucleotides were immobilized onto virgin glass microslides. Briefly, modified oligonucleotides were reacted first with 3-glycidyloxypropyltriethoxysilane (GOPTS), and subsequently, the resultant conjugates were directly immobilized onto the virgin glass surface by making use of silanization chemistry. The constructed microarrays were then used for discrimination of base mismatches. On subjecting to different pH and thermal conditions, the microarray showed sufficient stability. Application of this chemistry to manufacture oligonucleotide probe-based microarrays for detection of bacterial meningitis is demonstrated. Single-step reaction for the formation of conjugates with the commercially available reagent (GOPTS), omission of capping step and surface modification, and efficient immobilization of oligonucleotides onto the virgin glass surface are the key features of the proposed strategy.
Collapse
Affiliation(s)
- Dalip Sethi
- Institute of Genomics and Integrative Biology (CSIR), Delhi University Campus, Delhi, India
| | | | | | | |
Collapse
|
15
|
Pinto GR, Yoshioka FKN, Clara CA, Santos MJ, Almeida JRW, Burbano RR, Rey JA, Casartelli C. WRN Cys1367Arg SNP is not associated with risk and prognosis of gliomas in Southeast Brazil. J Neurooncol 2008; 90:253-8. [PMID: 18670736 DOI: 10.1007/s11060-008-9664-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Accepted: 07/22/2008] [Indexed: 10/21/2022]
Abstract
Werner syndrome (WS) is a premature aging disorder characterized by early onset of symptoms related to normal aging and by a high predisposition to various types of cancer, including gliomas. WS is caused by inherited recessive mutations in the WRN gene, which encodes a helicase considered a caretaker of the genome. Aiming to study the role of WRN Cys1367Arg in glioma susceptibility and oncologic prognosis of patients, we investigated the genotype distribution of this single nucleotide polymorphism in 94 glioma patients and 100 healthy subjects. Comparisons of genotype distributions and allele frequencies did not reveal any significant difference between the groups. Overall and disease-free survival rates were calculated, but no statistically significant difference was observed. Our data suggest that WRN Cys1367Arg SNP is not involved either in susceptibility to developing gliomas or in patient survival, at least in the Brazilian population.
Collapse
Affiliation(s)
- Giovanny R Pinto
- Human Genetics Laboratory, Federal University of Piauí, Av. São Sebastião 2819, 64202-020 Parnaíba, PI, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Prognostic molecular markers with no impact on decision-making: the paradox of gliomas based on a prospective study. Br J Cancer 2008; 98:1830-8. [PMID: 18506188 PMCID: PMC2410116 DOI: 10.1038/sj.bjc.6604378] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
This study assessed the prognostic value of several markers involved in gliomagenesis, and compared it with that of other clinical and imaging markers already used. Four-hundred and sixteen adult patients with newly diagnosed glioma were included over a 3-year period and tumour suppressor genes, oncogenes, MGMT and hTERT expressions, losses of heterozygosity, as well as relevant clinical and imaging information were recorded. This prospective study was based on all adult gliomas. Analyses were performed on patient groups selected according to World Health Organization histoprognostic criteria and on the entire cohort. The endpoint was overall survival, estimated by the Kaplan–Meier method. Univariate analysis was followed by multivariate analysis according to a Cox model. p14ARF, p16INK4A and PTEN expressions, and 10p 10q23, 10q26 and 13q LOH for the entire cohort, hTERT expression for high-grade tumours, EGFR for glioblastomas, 10q26 LOH for grade III tumours and anaplastic oligodendrogliomas were found to be correlated with overall survival on univariate analysis and age and grade on multivariate analysis only. This study confirms the prognostic value of several markers. However, the scattering of the values explained by tumour heterogeneity prevents their use in individual decision-making.
Collapse
|
17
|
Haapasalo J, Hilvo M, Nordfors K, Haapasalo H, Parkkila S, Hyrskyluoto A, Rantala I, Waheed A, Sly WS, Pastorekova S, Pastorek J, Parkkila AK. Identification of an alternatively spliced isoform of carbonic anhydrase XII in diffusely infiltrating astrocytic gliomas. Neuro Oncol 2008; 10:131-8. [PMID: 18322268 DOI: 10.1215/15228517-2007-065] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Carbonic anhydrase XII (CA XII) is a transmembrane enzyme that is associated with neoplastic growth. CA XII has been proposed to be involved in acidification of the extracellular milieu, creating an appropriate microenvironment for rapid tumor growth. Because RNA sequence databases have indicated that two isoforms of CA XII might exist in human tissues, and because alternatively spliced protein forms have been linked to aggressive behavior of cancer cells, we designed a study to evaluate the presence of the two forms of CA XII in diffuse astrocytomas, a tumor type known for its aggressive and often noncurable behavior. Reverse transcription PCR of tumor samples surprisingly revealed that CA XII present in diffuse astrocytomas is mainly encoded by a shorter mRNA variant. We further showed by Western blotting that anti-CA XII antibody recognized both isoforms in the glioblastoma cell lines, and we then evaluated the expression of CA XII in astrocytomas using immunohistochemistry and correlated the results with various clinicopathological and molecular factors. Of 370 diffusely infiltrating astrocytomas, 363 cases (98%) showed immunoreactions for CA XII. Importantly, CA XII expression correlated with poorer patient prognosis in univariate (p = 0.010, log-rank test) and multivariate survival analyses (p = 0.039, Cox analysis). From these results, we conclude that CA XII is commonly expressed in diffuse astrocytomas and that it might be used as a biomarker of poor prognosis. The absence of 11 amino acids in the shorter isoform, which seems to be common in astrocytomas, may affect the normal quaternary structure and biological function of CA XII.
Collapse
Affiliation(s)
- Joonas Haapasalo
- Institute of Medical Technology, University of Tampere, Biokatu 6, FI-33520 Tampere, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Stathopoulos A, Samuelson C, Milbouw G, Hermanne JP, Schijns VEJ, Chen TC. Therapeutic vaccination against malignant gliomas based on allorecognition and syngeneic tumor antigens: Proof of principle in two strains of rat. Vaccine 2008; 26:1764-72. [DOI: 10.1016/j.vaccine.2008.01.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Revised: 12/20/2007] [Accepted: 01/11/2008] [Indexed: 11/30/2022]
|
19
|
Mangiola A, Lama G, Giannitelli C, De Bonis P, Anile C, Lauriola L, La Torre G, Sabatino G, Maira G, Jhanwar-Uniyal M, Sica G. Stem Cell Marker Nestin and c-Jun NH2-Terminal Kinases in Tumor and Peritumor Areas of Glioblastoma Multiforme: Possible Prognostic Implications. Clin Cancer Res 2007; 13:6970-7. [DOI: 10.1158/1078-0432.ccr-07-1229] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Haque A, Banik NL, Ray SK. Emerging Role of Combination of All-trans Retinoic Acid and Interferon-gamma as Chemoimmunotherapy in the Management of Human Glioblastoma. Neurochem Res 2007; 32:2203-9. [PMID: 17676389 DOI: 10.1007/s11064-007-9420-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2007] [Accepted: 06/18/2007] [Indexed: 02/03/2023]
Abstract
Glioblastoma is the most malignant and common type of brain tumor with devastating outcome. Because current treatment modalities are mostly ineffective in controlling and curing glioblastoma, new and innovative therapeutic strategies must be developed. This article describes recent advances in chemoimmunotherapy, which is combination of chemotherapy and immunotherapy, against glioblastoma. We provide an overview of available treatment options for glioblastomas, gaps in our knowledge of immune recognition of these malignant tumors, and chemotherapeutic and immunotherapeutic agents that need to be further explored for designing novel chemoimmunotherapeutic strategy for the management of human glioblastomas. Our recent study demonstrated that combination of the chemotherapeutic agent all-trans retinoic acid (ATRA) and the immunotherapeutic agent interferon-gamma (IFN-gamma) could concurrently induce differentiation, apoptotic death, and immune components in two different human glioblastoma cell lines. We propose that combination of ATRA and IFN-gamma can become an efficacious chemoimmunotherapy for the treatment of human glioblastoma.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
21
|
Eoli M, Menghi F, Bruzzone MG, De Simone T, Valletta L, Pollo B, Bissola L, Silvani A, Bianchessi D, D'Incerti L, Filippini G, Broggi G, Boiardi A, Finocchiaro G. Methylation ofO6-Methylguanine DNA Methyltransferase and Loss of Heterozygosity on 19q and/or 17p Are Overlapping Features of Secondary Glioblastomas with Prolonged Survival. Clin Cancer Res 2007; 13:2606-13. [PMID: 17473190 DOI: 10.1158/1078-0432.ccr-06-2184] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Recent data suggest that methylation of the DNA repair gene O(6)-methylguanine DNA methyltransferase (MGMT), by increasing the chemosensitivity of glioblastoma multiforme, is significantly associated with improved prognosis. Results in contradiction with these findings, however, are present in the literature and the clinical and genetic context framing MGMT methylation is poorly characterized. EXPERIMENTAL DESIGN To address these issues, we have investigated the MGMT methylation status, clinical and magnetic resonance imaging characteristics, and relevant genetic features (loss of heterozygosity on 17p and 19q, EGFR amplification, and p53 mutations) in a retrospective study on 86 patients affected by glioblastoma multiforme: 72 patients had a clinical history indicating de novo insurgence of the tumor and the remaining 14 were secondary glioblastoma multiforme. RESULTS MGMT methylation was detected by methylation-specific PCR in 41 of 86 cases (47.7%; Meth+). Progression-free survival and overall survival were significantly longer in Meth+ than in Meth- patients [10 versus 7 months (P=0.003, log-rank test) and 18 versus 14 months (P=0.0003, log-rank test), respectively]. Mixed-nodular enhancement at magnetic resonance imaging was significantly more frequent in Meth+ and secondary glioblastoma multiforme and ring enhancement in Meth- and primary glioblastoma multiforme (P<0.005). MGMT methylation was more present in secondary glioblastoma multiforme (P=0.006) and associated with loss of heterozygosity on 17p and/or 19q (P=0.005). CONCLUSIONS These observations suggest that MGMT methylation is part of a genetic signature of glioblastomas that developed from lower-grade gliomas.
Collapse
Affiliation(s)
- Marica Eoli
- Unit of Clinical Neuro-Oncology, Istituto Nazionale Neurologico Besta, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cheng G, Zhang Y, Zhang X, Tang HF, Cao WD, Gao DK, Wang XL. Tubeimoside V (1), a new cyclic bisdesmoside from tubers of Bolbostemma paniculatum, functions by inducing apoptosis in human glioblastoma U87MG cells. Bioorg Med Chem Lett 2006; 16:4575-80. [PMID: 16784856 DOI: 10.1016/j.bmcl.2006.06.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Revised: 05/23/2006] [Accepted: 06/06/2006] [Indexed: 01/26/2023]
Abstract
Malignant glioblastoma is one of the most common malignant tumors in the neurological system. Tubeimoside V (1), a new cyclic bisdesmoside from tubers of Bolbostemma paniculatum, appears to exhibit various biological activities, including antitumor effect, but the function and mechanism of this new agent on glioblastoma cells has not previously been determined. In the present study, we investigated the proliferation change of human glioblastoma U87MG cells exposured to different concentrations (0.9-14.8 microM) of Tubeimoside V (1) for a certain time. The results showed that Tubeimoside V (1) significantly suppressed U87MG cell proliferation in a time- and dose-dependent manner (IC(50) = 3.6 microM). Flow cytometric analysis of DNA in U87MG cells showed that Tubeimoside V (1) induces the prominent appearance of a sub-G1 peak in the cell cycle suggestive of apoptosis. Furthermore, U87MG cells' treatment with Tubeimoside V (1) resulted in nuclear condensation with apoptotic bodies observed by both fluorescence and electron microscopy. The result of annexin V/PI assay showed that phosphatidylserine externalization began after treatment, and then increased in the following 24h. Molecular changes explored through Western-blot staining showed Tubeimoside V (1) decreased the expression levels of Bcl-2 protein and increased the expression levels of Bax protein. The novel findings suggest that the cytotoxic actions of Tubeimoside V (1) toward U87MG cells result from the induction of cell apoptosis. Overall, our data demonstrate that Tubeimoside V (1) is an efficient apoptotic killing agent of glioblastoma cells and suggest that this mechanism may play a critical role in anti-tumor chemotherapy.
Collapse
Affiliation(s)
- Guang Cheng
- Department of Neurosurgery of Xijing Hospital (Neurosurgery institute of Chinese PLA), The Fourth Military Medical University, Xi'an, ShannXi, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Le Jeune N, Dubois F, Bin V, Perek N. Evaluation of imatinib mesylate effects on glioblastoma aggressiveness with SPECT radiotracer 99mTc-(v)-DMSA. Eur J Cancer 2006; 42:1004-13. [PMID: 16564690 DOI: 10.1016/j.ejca.2006.01.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Revised: 12/19/2005] [Accepted: 01/03/2006] [Indexed: 11/20/2022]
Abstract
In vitro and in vivo studies have demonstrated inhibition of glioblastoma growth by imatinib mesylate (Gleevec). Imatinib is an inhibitor of the tyrosine kinase activities of platelet-derived growth factor receptor (PDGF-r), which is involved in glioblastoma aggressiveness. In this study, we have investigated the link between 99mTc-(V)-DMSA, an imaging agent used in Single Photon Emission Computed Tomography, cellular accumulation and the biological effects of imatinib mediated by PDGF-r in a human glioblastoma cell line U87-MG. Cells treated with imatinib showed significant decreases in proliferation, invasion, migration and PDGF-rbeta expression. 99mTc-(V)-DMSA cellular uptake studies showed that the specific action of imatinib on PDGF-r signal pathway, in the human glioblastoma cell line U87-MG, could be followed by radioactive tracer. Furthermore, strong correlations between cellular 99mTc-(V)-DMSA uptake and the effect of imatinib therapy on U87-MG proliferation (r=0.896), invasion (r=0.621) and migration (r=0.822) were obtained, likewise for 99mTc-(V)-DMSA uptake and PDGF-r expression (r=0.958). Our results show that the biological effects of imatinib therapy on tumour cells properties are linked to PDGF-r phosphorylation and could be traced with 99mTc-(V)-DMSA, which also seems to be a potential tracer to evaluate the response to imatinib therapy in glioblastoma.
Collapse
Affiliation(s)
- Nathalie Le Jeune
- Department of Biophysics and Radiopharmaceuticals, Research group EA 3063 "Cellular Survival and Adhesion in tumours and grafts", Faculty of Medicine Jacques Lisfranc, University of Saint-Etienne, France.
| | | | | | | |
Collapse
|
24
|
Cheng G, Zhang X, Tang HF, Zhang Y, Zhang XH, Cao WD, Gao DK, Wang XL, Jin BQ. Asterosaponin 1, a cytostatic compound from the starfish Culcita novaeguineae, functions by inducing apoptosis in human glioblastoma U87MG cells. J Neurooncol 2006; 79:235-41. [PMID: 16628475 DOI: 10.1007/s11060-006-9136-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Accepted: 02/09/2006] [Indexed: 11/30/2022]
Abstract
Malignant glioblastoma is one of the most common malignant tumors in the neurological system. Asterosaponin 1, a new cytostatic agent from the starfish Culcita novaeguineae appear to exhibit various biological activities, including antitumor effect, but the function and mechanism of this new agent on glioblastoma cells has not previously been determined. In the present study, we investigated the proliferation change of human glioblastoma U87MG cells exposed to different concentrations (2.5-20.0 microg/ml) of asterosaponin 1 for a certain time. The results showed that asterosaponin 1 significantly suppressed U87MG cell proliferation in a time- and dose-dependent manner (IC50 =4.3 microg/ml). Flow cytometric analysis of DNA in U87MG cells showed that asterosaponin 1 induces the prominent appearance of a sub-G1 peak in the cell cycle suggestive of apoptosis identical with the result of annexin V/PI assay. Furthermore, U87MG cells treatment with asterosaponin 1 resulted in nuclear condensation with apoptotic bodies observed by both fluorescence and electron microscopy. Agarose gel electrophoresis of DNA from asterosaponin 1-treated cells revealed a typical "ladder" consistent with apoptotic DNA fragmentation. Western-blot staining showed asterosaponin 1 decreased the expression of Bcl-2 protein and increased the expression of Bax protein. The novel findings suggest that the cytostatic actions of asterosaponin 1 toward U87MG cells result from the induction of cell apoptosis. Overall, our data demonstrate that asterosaponin 1 is fully equipped for an efficient apoptotic killing of glioblastoma cells and suggest that this mechanism may play a critical role in anti-tumor chemotherapy.
Collapse
Affiliation(s)
- Guang Cheng
- Department of Neurosurgery of Xijing Hospital (Neurosurgery institute of Chinese PLA), The Fourth Military Medical University, 710032, Xi'an, ShannXi, China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hou LC, Veeravagu A, Hsu AR, Tse VCK. Recurrent glioblastoma multiforme: a review of natural history and management options. Neurosurg Focus 2006; 20:E5. [PMID: 16709036 DOI: 10.3171/foc.2006.20.4.2] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive primary brain tumors, with a grim prognosis despite maximal treatment. Advancements in the past decades have not significantly increased the overall survival of patients with this disease. The recurrence of GBM is inevitable, its management often unclear and case dependent. In this report, the authors summarize the current literature regarding the natural history, surveillance algorithms, and treatment options of recurrent GBM. Furthermore, they provide brief discussions regarding current novel efforts in basic and clinical research. They conclude that although recurrent GBM remains a fatal disease, the literature suggests that a subset of patients may benefit from maximal treatment efforts. Nevertheless, further research effort in all aspects of GBM diagnosis and treatment remains essential to improve the overall prognosis of this disease.
Collapse
Affiliation(s)
- Lewis C Hou
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305-5327, USA
| | | | | | | |
Collapse
|