1
|
Checa-Chavarria E, Rivero-Buceta E, Sanchez Martos MA, Martinez Navarrete G, Soto-Sánchez C, Botella P, Fernández E. Development of a Prodrug of Camptothecin for Enhanced Treatment of Glioblastoma Multiforme. Mol Pharm 2021; 18:1558-1572. [PMID: 33645231 PMCID: PMC8482753 DOI: 10.1021/acs.molpharmaceut.0c00968] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
A novel therapeutic approach for
glioblastoma multiforme (GBM)
therapy has been carried out through in vitro and in vivo testing by using the prodrug camptothecin-20-O-(5-aminolevulinate) (CPT-ALA). The incorporation of ALA
to CPT may promote uptake of the cytotoxic molecule by glioblastoma
cells where the heme synthesis pathway is active, improving the therapeutic
action and reducing the side effects over healthy tissue. The antitumor
properties of CPT-ALA have been tested on different GBM cell lines
(U87, U251, and C6) as well as in an orthotopic GBM model in rat,
where potential toxicity in central nervous system cells was analyzed. In vitro results indicated no significant differences in
the cytotoxic effect over the different GBM cell lines for CPT and
CPT-ALA, albeit cell mortality induced by CPT over normal cell lines
was significantly higher than CPT-ALA. Moreover, intracranial GBM
in rat was significantly reduced (30% volume) with 2 weeks of CPT-ALA
treatment with no significant side effects or alterations to the well-being
of the animals tested. 5-ALA moiety enhances CPT diffusion into tumors
due to solubility improvement and its metabolic-based targeting, increasing
the CPT cytotoxic effect on malignant cells while reducing CPT diffusion
to other proliferative healthy tissue. We demonstrate that CPT-ALA
blocks proliferation of GBM cells, reducing the infiltrative capacity
of GBM and promoting the success of surgical removal, which improves
life expectancy by reducing tumor recurrence.
Collapse
Affiliation(s)
- Elisa Checa-Chavarria
- Institute of Bioengineering, Universidad Miguel Hernández, Elche, Spain and Centre for Network Biomedical Research (CIBER-BBN), Avenida de la Universidad s/n, 03202 Elche, Spain
| | - Eva Rivero-Buceta
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, Avenida de los Naranjos s/n, 46022 Valencia, Spain
| | - Miguel Angel Sanchez Martos
- Institute of Bioengineering, Universidad Miguel Hernández, Elche, Spain and Centre for Network Biomedical Research (CIBER-BBN), Avenida de la Universidad s/n, 03202 Elche, Spain
| | - Gema Martinez Navarrete
- Institute of Bioengineering, Universidad Miguel Hernández, Elche, Spain and Centre for Network Biomedical Research (CIBER-BBN), Avenida de la Universidad s/n, 03202 Elche, Spain
| | - Cristina Soto-Sánchez
- Institute of Bioengineering, Universidad Miguel Hernández, Elche, Spain and Centre for Network Biomedical Research (CIBER-BBN), Avenida de la Universidad s/n, 03202 Elche, Spain
| | - Pablo Botella
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, Avenida de los Naranjos s/n, 46022 Valencia, Spain
| | - Eduardo Fernández
- Institute of Bioengineering, Universidad Miguel Hernández, Elche, Spain and Centre for Network Biomedical Research (CIBER-BBN), Avenida de la Universidad s/n, 03202 Elche, Spain
| |
Collapse
|
2
|
Abstract
New discoveries in Glioblastoma (GBM) biology have been made using genomics data. Genomic markers are routinely integrated into clinical neurosurgical practice. In this manuscript, we review the fundamentals of genomics such as the differences between first, second, and third generation sequencing technology. We also review the impact of single cell genomics in understanding the complex heterogenous GBM microenvironment. Finally, we will discuss advances in epigenetics that have lent insights into treatment resistance. The integration of genomics into neuro-oncology clinical practice is routine and will continue to expand with the expansion of precision of medicine. We provide a primer for clinicians.
Collapse
Affiliation(s)
- John D Patterson
- Department of Neurological Surgery, University of Arkansas for Medical Sciences
| | | | - Analiz Rodriguez
- Department of Neurological Surgery, University of Arkansas for Medical Sciences
| |
Collapse
|
3
|
Ius T, Pignotti F, Della Pepa GM, La Rocca G, Somma T, Isola M, Battistella C, Gaudino S, Polano M, Dal Bo M, Bagatto D, Pegolo E, Chiesa S, Arcicasa M, Olivi A, Skrap M, Sabatino G. A Novel Comprehensive Clinical Stratification Model to Refine Prognosis of Glioblastoma Patients Undergoing Surgical Resection. Cancers (Basel) 2020; 12:E386. [PMID: 32046132 PMCID: PMC7072471 DOI: 10.3390/cancers12020386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 12/14/2022] Open
Abstract
Despite recent discoveries in genetics and molecular fields, glioblastoma (GBM) prognosis still remains unfavorable with less than 10% of patients alive 5 years after diagnosis. Numerous studies have focused on the research of biological biomarkers to stratify GBM patients. We addressed this issue in our study by using clinical/molecular and image data, which is generally available to Neurosurgical Departments in order to create a prognostic score that can be useful to stratify GBM patients undergoing surgical resection. By using the random forest approach [CART analysis (classification and regression tree)] on Survival time data of 465 cases, we developed a new prediction score resulting in 10 groups based on extent of resection (EOR), age, tumor volumetric features, intraoperative protocols and tumor molecular classes. The resulting tree was trimmed according to similarities in the relative hazard ratios amongst groups, giving rise to a 5-group classification tree. These 5 groups were different in terms of overall survival (OS) (p < 0.000). The score performance in predicting death was defined by a Harrell's c-index of 0.79 (95% confidence interval [0.76-0.81]). The proposed score could be useful in a clinical setting to refine the prognosis of GBM patients after surgery and prior to postoperative treatment.
Collapse
Affiliation(s)
- Tamara Ius
- Neurosurgery Unit, Department of Neuroscience, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Fabrizio Pignotti
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
| | | | - Giuseppe La Rocca
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Università degli Studi di Napoli Federico II, 80131 Naples, Italy;
| | - Miriam Isola
- Department of Medicine, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (M.I.); (C.B.)
| | - Claudio Battistella
- Department of Medicine, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (M.I.); (C.B.)
| | - Simona Gaudino
- Institute of radiology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (M.P.); (M.D.B.)
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (M.P.); (M.D.B.)
| | - Daniele Bagatto
- Neuroradiology Unit, Department of Diagnostic Imaging ASUIUD Udine, 33100 Udine, Italy;
| | - Enrico Pegolo
- Institute of Pathology, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Silvia Chiesa
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Mauro Arcicasa
- Department of Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| | - Alessandro Olivi
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| | - Miran Skrap
- Neurosurgery Unit, Department of Neuroscience, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Giovanni Sabatino
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| |
Collapse
|
4
|
Ius T, Pignotti F, Della Pepa GM, Bagatto D, Isola M, Battistella C, Gaudino S, Pegolo E, Chiesa S, Arcicasa M, La Rocca G, Olivi A, Skrap M, Sabatino G. Glioblastoma: from volumetric analysis to molecular predictors. J Neurosurg Sci 2020; 66:173-186. [PMID: 32031360 DOI: 10.23736/s0390-5616.20.04850-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Despite decades of therapeutic and molecular refinements, the prognosis of patients with glioblastoma (GBM) still remains unfavorable. Integrative clinical studies allow a better understanding of the natural evolution of GBM. To assess independent predictors of overall survival (OS) and progression free survival (PFS) clinical, surgical, molecular and radiological variables were evaluated. A novel preoperative volumetric magnetic resonance imaging (MRI) index for tumor prognosis in GBM patients was investigated. METHODS A cohort of 195 cases of patients operated for newly GBM were analyzed. Extent of tumoral resection (EOR), tumor growth pattern, expressed by preoperative volumetric ΔT1-T2 MRI index, molecular markers such as O6-methylguanine-DNA methyltransferase (MGMT) methylation and isocitrate dehydrogenase 1/2 (IDH1/2) mutation, were analyzed. Analysis of survival was done using Cox-proportional hazard models. RESULTS The 1-, 2- years estimated OS and PFS rate for the whole population were 61% and 27%, 38% and 17%, respectively. A better survival rate, both in terms of survival and tumor progression, was observed in patient with higher EOR (p=0.000), younger age (p=0.000), MGMT methylation status (p=0.001) and lower preoperative ΔT1-T2 MRI index (p=0.004). Regarding the tumor growth pattern a cut-off value of 0.75 was found to discriminate patient with different prognosis. Patients with a preoperative ΔT1-T2 MRI index <0.75 had a 1-year estimated OS of 67%, otherwise patients with a preoperative ΔT1-T2 MRI index >0.75 hada 1-year estimated OS of 34%. CONCLUSIONS In this investigation longer survival is associated with younger age, EOR, promoter methylation of MGMT and preoperative tumor volumetric features expressed by ΔT1-T2 MRI index. The preoperative ΔT1-T2 MRI index could be a promising prognostic factor potentially useful in GBM management. Future investigations based on multiparametric MRI data and next generation sequences analysis, may better clarify this result.
Collapse
Affiliation(s)
- Tamara Ius
- Neurosurgery Unit, Department of Neuroscience, Santa Maria della Misericordia University Hospital, Udine, Italy -
| | | | | | - Daniele Bagatto
- Neuroradiology Unit, Department of Diagnostic Imaging ASUIUD Udine, Italy
| | - Miriam Isola
- Department of Medicine, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Claudio Battistella
- Department of Medicine, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Simona Gaudino
- Institute of Radiology, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Enrico Pegolo
- Institute of Pathology, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Silvia Chiesa
- Institute of Radiotherapy, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | | | | | | | - Miran Skrap
- Neurosurgery Unit, Department of Neuroscience, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Giovanni Sabatino
- Institute of Neurosurgery, Catholic University, Rome, Italy.,Department of Neurosurgery, Mater Olbia Hospital, Olbia, Italy
| |
Collapse
|
5
|
Identification of miRNA signatures in serum exosomes as a potential biomarker after radiotherapy treatment in glioma patients. Ann Diagn Pathol 2019; 44:151436. [PMID: 31865249 DOI: 10.1016/j.anndiagpath.2019.151436] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Despite development in therapies, the high recurrence and low positivity of biomarkers for diagnosis still result in glioma with high mortality. In this study, we aimed to identify a potential miRNA signature to evaluate the effect of radiotherapy in glioma patients. MicroRNA (miRNA) sequencing was performed on miRNAs isolated from serum exosomes in a cohort of glioma patients before and after radiotherapy. A total of 18 up-regulated differentially expressed (DE) miRNAs and 16 down-regulated DE miRNAs were identified. Subsequently, the target genes of DE miRNAs were predicted based on multiple miRNA-target databases. Further, it was indicated that these targets were primarily involved in metabolic process, p53 signaling pathway and cancer pathways, suggesting that these miRNAs play a crucial role in glioma by regulating targets and affect the occurrence and development of the disease. In general, this study presented the variation of miRNAs in blood exosomes before and after radiotherapy. It can not only be helpful for the diagnosis of glioma, but also find new candidate biomarkers for monitoring the condition and evaluating the efficacy of radiotherapy in glioma. It provides a new idea for the diagnosis, treatment and prognosis evaluation of glioma.
Collapse
|
6
|
He X, Zhang S, Chen J, Li D. Increased LGALS3 expression independently predicts shorter overall survival in patients with the proneural subtype of glioblastoma. Cancer Med 2019; 8:2031-2040. [PMID: 30848102 PMCID: PMC6536958 DOI: 10.1002/cam4.2075] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/05/2019] [Accepted: 02/15/2019] [Indexed: 11/24/2022] Open
Abstract
In the current study, we tried to study the expression of LGALS3 and LGALS3BP, their potential as prognostic markers and the possible genetic/epigenetic mechanisms underlying their dysregulation in different subtypes of glioblastoma (GBM). An in silico retrospective study was performed using large online databases. Results showed that LGALS3 and LGALS3BP were upregulated at both RNA and protein levels in GBM tissue and were generally associated with shorter overall survival (OS) in GBM patients. However, in subgroup analysis, we only found the association in proneural subtype. The copy number alterations did not necessarily lead to LGALS3/LGALS3BP dysregulation. In the proneural subtype of GBM patients, hypermethylation of the two CpG sites (cg19099850 and cg17403875) was associated with significantly lower expression of LGALS3. In univariate and multivariate analysis, LGALS3 expression independently predicted shorter OS in the proneural subtype of GBM (HR: 1.487, 95% CI: 1.229‐1.798, P < 0.001), after adjustment of age, gender, IDH1 mutations, temozolomide chemotherapy, radiotherapy and LGALS3BP expression. In comparison, LGALS3BP lost the prognostic value in multivariate analysis. Based on these findings, we infer that LGALS3 expression serves as an independent biomarker of shorter OS in the proneural subtype of GBM, the expression of which might be regulated in an epigenetic manner.
Collapse
Affiliation(s)
- Xia He
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Bayi Rehabilitation Center/Sichuan Provincial Rehabilitation Hospital, Chengdu, China
| | - Sunfu Zhang
- Department of Neurosurgery, The First People's Hospital of Yibin, Yibin, China
| | - Junchen Chen
- Department of Neurosurgery, Sichuan Bayi Rehabilitation Center/Sichuan Provincial Rehabilitation Hospital, Chengdu, China
| | - Dekang Li
- Department of Neurosurgery, The First People's Hospital of Yibin, Yibin, China
| |
Collapse
|
7
|
Hu JL, Luo WJ, Wang H. Angiogenin Upregulation Independently Predicts Unfavorable Overall Survival in Proneural Subtype of Glioblastoma. Technol Cancer Res Treat 2019; 18:1533033819846636. [PMID: 31072237 PMCID: PMC6515846 DOI: 10.1177/1533033819846636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Objective: Angiogenin is a small protein that exerts potent stimulating effects on angiogenesis. In this study, we aimed to examine the expression of angiogenin in different subtypes of glioblastoma and estimated its independent prognostic value. Methods: The genomic and survival data from The Cancer Genome Atlas-glioblastoma were extracted for a secondary study. Results The expression of angiogenin was upregulated in glioblastoma tissues and varied significantly in different subtypes. Although the proneural subtype had the lowest angiogenin expression, high angiogenin expression was associated with significantly worse overall survival. However, this association was not observed in other subtypes. By performing univariate and multivariate analysis using Cox regression model, we observed that high angiogenin expression was an independent indicator of shorter overall survival in proneural glioblastoma (hazard ratio: 1.669, 95% confidence interval: 1.033-2.696, P = .036), after adjustment of age, gender, isocitrate dehydrogenase 1 mutation, temozolomide chemotherapy and radiation therapy. In addition, we also observed a correlation between elevated angiogenin expression and the hypomethylated status of its DNA. The hypermethylation group had significantly better overall survival. Conclusions: Angiogenin upregulation might serve as a biomarker for unfavorable overall survival in the proneural subtype of glioblastoma.
Collapse
Affiliation(s)
- Ji-Liang Hu
- 1 Department of Neurosurgery, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Wei-Jian Luo
- 1 Department of Neurosurgery, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Hao Wang
- 1 Department of Neurosurgery, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| |
Collapse
|
8
|
Chen Z, Wei X, Shen L, Zhu H, Zheng X. 20(S)-ginsenoside-Rg3 reverses temozolomide resistance and restrains epithelial-mesenchymal transition progression in glioblastoma. Cancer Sci 2018; 110:389-400. [PMID: 30431207 PMCID: PMC6317960 DOI: 10.1111/cas.13881] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/11/2018] [Accepted: 11/12/2018] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant human intracranial tumors. Temozolomide (TMZ) is the primary alkylating agent for GBM patients. However, many GBM patients are resistant to TMZ. Therefore, patients with GBM urgently need more effective therapeutic options. 20(S)‐ginsenoside‐Rg3 (20(S)‐Rg3) is a natural chemical with anti‐tumor effects, but at present there is little understanding of its functional mechanism. Several research reports have demonstrated that O6‐methylguanine DNA‐methyltransferase (MGMT) repairs damaged DNA and contributes to TMZ resistance in gliomas. In addition, recent studies have shown that MGMT gene expression could be regulated by the Wnt/β‐catenin pathway. However, whether 20(S)‐Rg3 inhibits MGMT expression and augments chemosensitivity to Temozolomide (TMZ) in glioma cells remains unclear. In this study, we explored the modulating effects of 20(S)‐Rg3 on MGMT. We used glioma cell lines, primary cell strain (including T98G, U118 and GBM‐XX; all of them are MGMT‐positive glioma cell lines) and xenograft glioma models to examine whether 20(S)‐Rg3 increased the sensitivity to TMZ and to reveal the underlying mechanisms. We found that the MGMT expression was effectively downregulated by 20(S)‐Rg3 via the Wnt/β‐catenin pathway in glioma cell lines, and TMZ resistance was significantly reversed by 20(S)‐Rg3. Meanwhile, 20(S)‐Rg3 shows no obvious cytotoxicity at its effective dose and is well tolerated in vivo. In addition, we found that 20(S)‐Rg3 significantly restrains the epithelial‐mesenchymal transition (EMT) progression of glioma cells. Taken together, these results indicate that 20(S)‐Rg3 may be a novel agent to use in treatment of GBM, especially in TMZ‐resistant GBM with high MGMT expression.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiangyu Wei
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lin Shen
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Hanshuo Zhu
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xuesheng Zheng
- Department of Neurosurgery, XinHua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Khan UA, Rennert RC, White NS, Bartsch H, Farid N, Dale AM, Chen CC. Diagnostic utility of restriction spectrum imaging (RSI) in glioblastoma patients after concurrent radiation-temozolomide treatment: A pilot study. J Clin Neurosci 2018; 58:136-141. [PMID: 30253908 DOI: 10.1016/j.jocn.2018.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 05/01/2018] [Accepted: 09/10/2018] [Indexed: 01/21/2023]
Abstract
Discriminating between tumor recurrence and treatment effects in glioblastoma patients undergoing radiation-temozolomide (RT/TMZ) therapy remains a major clinical challenge. Here, we report a pilot study to determine the utility of restriction spectrum imaging (RSI), an advanced diffusion-weighted MRI (DWI) technique that affords meso-scale resolution of cell density, in this assessment. A retrospective review of 31 patients with glioblastoma treated between 2011 and 2017 who underwent surgical resection or biopsy over radiographic concern for tumor recurrence following RT/TMZ was performed. All patients underwent RSI prior to surgical resection. Diagnostic utility of RSI for tumor recurrence was determined in comparison to histopathology. Analysis of surgical specimens revealed treatment effects in 6/31 patients (19%) and tumor recurrence in 25/31 patients (81%). There was general concordance between the measured RSI signal and histopathologic diagnosis. RSI was negative in 5/6 patients (83%) in patients with histological evidence of treatment effects. RSI was positive in 21/25 patients (84%) in patients with tumor recurrence. The sensitivity, specificity, positive and negative predictive values of RSI for glioblastoma recurrence were 84%, 86%, 95%, and 60%, respectively. Histopathologic review showed agreement between the RSI signal and cellularity of the tumor specimen. These data support the use of RSI in the evaluation of treatment effects versus tumor recurrence in glioblastoma patients after RT-TMZ therapy.
Collapse
Affiliation(s)
- Usman A Khan
- Department of Neurosurgery, University of California San Diego, San Diego, CA, USA
| | - Robert C Rennert
- Department of Neurosurgery, University of California San Diego, San Diego, CA, USA
| | - Nathan S White
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
| | - Hauke Bartsch
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
| | - Nikdokht Farid
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA; Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
10
|
Treiber JM, Steed TC, Brandel MG, Patel KS, Dale AM, Carter BS, Chen CC. Molecular physiology of contrast enhancement in glioblastomas: An analysis of The Cancer Imaging Archive (TCIA). J Clin Neurosci 2018; 55:86-92. [PMID: 29934058 DOI: 10.1016/j.jocn.2018.06.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 03/06/2018] [Accepted: 06/04/2018] [Indexed: 12/23/2022]
Abstract
The physiologic processes underlying MRI contrast enhancement in glioblastoma patients remain poorly understood. MRIs of 148 glioblastoma subjects from The Cancer Imaging Archive were segmented using Iterative Probabilistic Voxel Labeling (IPVL). Three aspects of contrast enhancement (CE) were parametrized: the mean intensity of all CE voxels (CEi), the intensity heterogeneity in CE (CEh), and volumetric ratio of CE to necrosis (CEr). Associations between these parameters and patterns of gene expression were analyzed using DAVID functional enrichment analysis. Glioma CpG island methylator phenotype (G-CIMP) glioblastomas were poorly enhancing. Otherwise, no differences in CE parameters were found between proneural, neural, mesenchymal, and classical glioblastomas. High CEi was associated with expression of genes that mediate inflammatory responses. High CEh was associated with increased expression of genes that regulate remodeling of extracellular matrix (ECM) and endothelial permeability. High CEr was associated with increased expression of genes that mediate cellular response to stressful metabolic states, including hypoxia and starvation. Our results indicate that CE in glioblastoma is associated with distinct biological processes involved in inflammatory response and tissue hypoxia. Integrative analysis of these CE parameters may yield meaningful information pertaining to the biologic state of glioblastomas and guide future therapeutic paradigms.
Collapse
Affiliation(s)
- Jeffrey M Treiber
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA.
| | - Tyler C Steed
- Department of Neurosurgery, Emory University, Atlanta, GA, USA.
| | - Michael G Brandel
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA, USA.
| | - Kunal S Patel
- Department of Neurosurgery, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA; Department of Radiology, University of California San Diego, La Jolla, CA, USA.
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA.
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
11
|
Alattar AA, Brandel MG, Hirshman BR, Dong X, Carroll KT, Ali MA, Carter BS, Chen CC. Oligodendroglioma resection: a Surveillance, Epidemiology, and End Results (SEER) analysis. J Neurosurg 2018; 128:1076-1083. [DOI: 10.3171/2016.11.jns161974] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVEThe available evidence suggests that the clinical benefits of extended resection are limited for chemosensitive tumors, such as primary CNS lymphoma. Oligodendroglioma is generally believed to be more sensitive to chemotherapy than astrocytoma of comparable grades. In this study the authors compare the survival benefit of gross-total resection (GTR) in patients with oligodendroglioma relative to patients with astrocytoma.METHODSUsing the Surveillance, Epidemiology, and End Results (SEER) Program (1999–2010) database, the authors identified 2378 patients with WHO Grade II oligodendroglioma (O2 group) and 1028 patients with WHO Grade III oligodendroglioma (O3 group). Resection was defined as GTR, subtotal resection, biopsy only, or no resection. Kaplan-Meier and multivariate Cox regression survival analyses were used to assess survival with respect to extent of resection.RESULTSCox multivariate analysis revealed that the hazard of dying from O2 and O3 was comparable between patients who underwent biopsy only and GTR (O2: hazard ratio [HR] 1.06, 95% confidence interval [CI] 0.73–1.53; O3: HR 1.18, 95% CI 0.80–1.72). A comprehensive search of the published literature identified 8 articles without compelling evidence that GTR is associated with improved overall survival in patients with oligodendroglioma.CONCLUSIONSThis SEER-based analysis and review of the literature suggest that GTR is not associated with improved survival in patients with oligodendroglioma. This finding contrasts with the documented association between GTR and overall survival in anaplastic astrocytoma and glioblastoma. The authors suggest that this difference may reflect the sensitivity of oligodendroglioma to chemotherapy as compared with astrocytomas.
Collapse
Affiliation(s)
| | | | - Brian R. Hirshman
- 2Division of Neurological Surgery, University of California, San Diego, California; and
- 3Computation, Organization, and Society Program, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | | | | | | | - Bob S. Carter
- 2Division of Neurological Surgery, University of California, San Diego, California; and
| | - Clark C. Chen
- 2Division of Neurological Surgery, University of California, San Diego, California; and
| |
Collapse
|
12
|
Lanman TA, Compton JN, Carroll KT, Hirshman BR, Ali MA, Lochte B, Carter B, Chen CC. Survival patterns of oligoastrocytoma patients: A surveillance, epidemiology and end results (SEER) based analysis. INTERDISCIPLINARY NEUROSURGERY 2018. [DOI: 10.1016/j.inat.2017.07.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
13
|
Steed TC, Treiber JM, Patel K, Ramakrishnan V, Merk A, Smith AR, Carter BS, Dale AM, Chow LML, Chen CC. Differential localization of glioblastoma subtype: implications on glioblastoma pathogenesis. Oncotarget 2018; 7:24899-907. [PMID: 27056901 PMCID: PMC5041878 DOI: 10.18632/oncotarget.8551] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/26/2016] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION The subventricular zone (SVZ) has been implicated in the pathogenesis of glioblastoma. Whether molecular subtypes of glioblastoma arise from unique niches of the brain relative to the SVZ remains largely unknown. Here, we tested whether these subtypes of glioblastoma occupy distinct regions of the cerebrum and examined glioblastoma localization in relation to the SVZ. METHODS Pre-operative MR images from 217 glioblastoma patients from The Cancer Imaging Archive were segmented automatically into contrast enhancing (CE) tumor volumes using Iterative Probabilistic Voxel Labeling (IPVL). Probabilistic maps of tumor location were generated for each subtype and distances were calculated from the centroid of CE tumor volumes to the SVZ. Glioblastomas that arose in a Genetically Modified Murine Model (GEMM) model were also analyzed with regard to SVZ distance and molecular subtype. RESULTS Classical and mesenchymal glioblastomas were more diffusely distributed and located farther from the SVZ. In contrast, proneural and neural glioblastomas were more likely to be located in closer proximity to the SVZ. Moreover, in a GFAP-CreER; PtenloxP/loxP; Trp53loxP/loxP; Rb1loxP/loxP; Rbl1-/- GEMM model of glioblastoma where tumor can spontaneously arise in different regions of the cerebrum, tumors that arose near the SVZ were more likely to be of proneural subtype (p < 0.0001). CONCLUSIONS Glioblastoma subtypes occupy different regions of the brain and vary in proximity to the SVZ. These findings harbor implications pertaining to the pathogenesis of glioblastoma subtypes.
Collapse
Affiliation(s)
- Tyler C Steed
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jeffrey M Treiber
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Kunal Patel
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Weill-Cornell Medical College, New York Presbyterian Hospital, New York, NY, USA
| | - Valya Ramakrishnan
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Alexander Merk
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Amanda R Smith
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bob S Carter
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA.,Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Lionel M L Chow
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Clark C Chen
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
14
|
Abstract
Glioblastoma multiforme (GBM) is the most lethal primary brain tumor in adults despite contemporary gold-standard first-line treatment strategies. This type of tumor recurs in virtually all patients and no commonly accepted standard treatment exists for the recurrent disease. Therefore, advances in all scientific and clinical aspects of GBM are urgently needed. Epigenetic mechanisms are one of the major factors contributing to the pathogenesis of cancers, including glioblastoma. Epigenetic modulators that regulate gene expression by altering the epigenome and non-histone proteins are being exploited as therapeutic drug targets. Over the last decade, numerous preclinical and clinical studies on histone deacetylase (HDAC) inhibitors have shown promising results in various cancers. This article provides an overview of the anticancer mechanisms of HDAC inhibitors and the role of HDAC isoforms in GBM. We also summarize current knowledge on HDAC inhibitors on the basis of preclinical studies and emerging clinical data.
Collapse
|
15
|
Carroll KT, Hirshman B, Ali MA, Alattar AA, Brandel MG, Lochte B, Lanman T, Carter B, Chen CC. Management and Survival Patterns of Patients with Gliomatosis Cerebri: A SEER-Based Analysis. World Neurosurg 2017; 103:186-193. [DOI: 10.1016/j.wneu.2017.03.103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/20/2017] [Accepted: 03/21/2017] [Indexed: 01/27/2023]
|
16
|
Akers JC, Hua W, Li H, Ramakrishnan V, Yang Z, Quan K, Zhu W, Li J, Figueroa J, Hirshman BR, Miller B, Piccioni D, Ringel F, Komotar R, Messer K, Galasko DR, Hochberg F, Mao Y, Carter BS, Chen CC. A cerebrospinal fluid microRNA signature as biomarker for glioblastoma. Oncotarget 2017; 8:68769-68779. [PMID: 28978155 PMCID: PMC5620295 DOI: 10.18632/oncotarget.18332] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/19/2017] [Indexed: 12/22/2022] Open
Abstract
Purpose To develop a cerebrospinal fluid (CSF) miRNA diagnostic biomarker for glioblastoma. Experimental Design Glioblastoma tissue and matched CSF from the same patient (obtained prior to tumor manipulation) were profiled by TaqMan OpenArray® Human MicroRNA Panel. CSF miRNA profiles from glioblastoma patients and controls were created from three discovery cohorts and confirmed in two validation cohorts. Results miRNA profiles from clinical CSF correlated with those found in glioblastoma tissues. Comparison of CSF miRNA profiles between glioblastoma patients and non-brain tumor patients yielded a tumor “signature” consisting of nine miRNAs. The “signature” correlated with glioblastoma tumor volume (p=0.008). When prospectively applied to cisternal CSF, the sensitivity and specificity of the ‘signature’ for glioblastoma detection were 67% and 80%, respectively. For lumbar CSF, the sensitivity and specificity of the signature were 28% and 95%, respectively. Comparable results were obtained from analyses of CSF extracellular vesicles (EVs) and crude CSF. Conclusion We report a CSF miRNA signature as a “liquid biopsy” diagnostic platform for glioblastoma.
Collapse
Affiliation(s)
- Johnny C Akers
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongying Li
- Biostatistics Department, Moores Cancer Center, UC San Diego Health System, La Jolla, CA, USA
| | - Valya Ramakrishnan
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Zixiao Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Kai Quan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Li
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Javier Figueroa
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Brian R Hirshman
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Brittney Miller
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - David Piccioni
- Department of Neurosurgery, Moores Cancer Center, UC San Diego Health System, La Jolla, CA, USA
| | - Florian Ringel
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Ricardo Komotar
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Karen Messer
- Biostatistics Department, Moores Cancer Center, UC San Diego Health System, La Jolla, CA, USA
| | - Douglas R Galasko
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Fred Hochberg
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Ying Mao
- Department of Neurosciences, University of California, San Diego, CA, USA.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Bob S Carter
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Clark C Chen
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| |
Collapse
|
17
|
Liu X, Chen J, Zhang J. AdipoR1-mediated miR-3908 inhibits glioblastoma tumorigenicity through downregulation of STAT2 associated with the AMPK/SIRT1 pathway. Oncol Rep 2017; 37:3387-3396. [PMID: 28440504 DOI: 10.3892/or.2017.5589] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 04/07/2017] [Indexed: 11/06/2022] Open
Abstract
A prospective method of treatment for cancer is to inhibit oncogene signaling pathways with microRNA (miRNA or miR). In the present study, whether the expression of STAT2, AdipoR1/AMPK/SIRT1 pathway of glioma is regulated by miR-3908 was explored. To confirm whether the predicted miR-3908 is matched with STAT2 and AdipoR1, 3'UTR luciferase activity of STAT2 and AdipoR1 was assessed. In the presence of the mimics or inhibitors of miR-3908, cell function of glioma cells, such as proliferation, growth, migration, invasion and apoptosis were analyzed. The expression of AdipoR1 and its downstream AMPK/SIRT1 pathway proteins or STAT2 were examined. Luciferase reporter analysis showed that miR-3908 directly target STAT2 and AdipoR1. miR-3908 suppressed expression of STAT2 or AdipoR1 and downregulated AdipoR1 pathway genes, including AMPK, p-AMPK and SIRT1. miR-3908 inhibited tumorigenicity, migration, growth and invasion in glioma cell lines U251 and U87 as well as increased apoptosis of these cells. The pathways related to tumorigenicity and tumor progression, STAT2 and AdipoR1/AMPK/SIRT1 could be restrained by miR-3908. In conclusion, restoration of miR-3908 expression induced suppression of cancer progression and glioblastoma tumorigenicity. The present study discovered novel tumorigenesis associated with miR-3908, which may represent a new target in treatment for glioblastoma.
Collapse
Affiliation(s)
- Xiangming Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Jinglong Chen
- Department of Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| | - Jinqian Zhang
- Department of Laboratory Medicine, The Second People's Hospital of Guangdong Province, Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| |
Collapse
|
18
|
Brandel MG, Alattar AA, Hirshman BR, Dong X, Carroll KT, Ali MA, Carter BS, Chen CC. Survival trends of oligodendroglial tumor patients and associated clinical practice patterns: a SEER-based analysis. J Neurooncol 2017; 133:173-181. [DOI: 10.1007/s11060-017-2430-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
|
19
|
Akers JC, Ramakrishnan V, Yang I, Hua W, Mao Y, Carter BS, Chen CC. Optimizing preservation of extracellular vesicular miRNAs derived from clinical cerebrospinal fluid. Cancer Biomark 2017; 17:125-32. [PMID: 27062568 DOI: 10.3233/cbm-160609] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Tumor specific genetic material can be detected in extracellular vesicles (EVs) isolated from blood, cerebrospinal fluid (CSF), and other biofluids of glioblastoma patients. As such, EVs have emerged as a promising platform for biomarker discovery. However, the optimal procedure to transport clinical EV samples remains poorly characterized. METHODS We examined the stability of EVs isolated from CSF of glioblastoma patients that were stored under different conditions. EV recovery was determined by Nanoparticle tracking analysis, and qRT-PCR was performed to determine the levels of miRNAs. RESULTS CSF EVs that were lyophilized and stored at room temperature (RT) for seven days exhibited a 37-43% reduction in EV number. This reduction was further associated with decreased abundance of representative miRNAs. In contrast, the EV number and morphology remained largely unchanged if CSF were stored at RT. Total RNA and representative miRNA levels were well-preserved under this condition for up to seven days. A single cycle of freezing and thawing did not significantly alter EV number, morphology, RNA content, or miRNA levels. However, incremental decreases in these parameters were observed after two cycles of freezing and thawing. CONCLUSIONS These results suggest that EVs in CSF are stable at RT for at least seven days. Repeated cycles of freezing/thawing should be avoided to minimize experimental artifacts.
Collapse
Affiliation(s)
- Johnny C Akers
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Valya Ramakrishnan
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Isaac Yang
- Department of Neurosurgery, University of California, Los Angeles, CA, USA
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Bob S Carter
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA.,Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Clark C Chen
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA.,Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| |
Collapse
|
20
|
Hirshman BR, Jones LA, Carroll KT, Tang JA, Proudfoot JA, Carley KM, Carter BS, Chen CC. Coevolution of Peer-Reviewed Literature and Clinical Practice in High-Grade Glioma Resection. World Neurosurg 2016; 96:237-241. [DOI: 10.1016/j.wneu.2016.07.105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 07/27/2016] [Accepted: 07/30/2016] [Indexed: 11/16/2022]
|
21
|
Yang P, Zhang W, Wang Y, Peng X, Chen B, Qiu X, Li G, Li S, Wu C, Yao K, Li W, Yan W, Li J, You Y, Chen CC, Jiang T. IDH mutation and MGMT promoter methylation in glioblastoma: results of a prospective registry. Oncotarget 2016; 6:40896-906. [PMID: 26503470 PMCID: PMC4747376 DOI: 10.18632/oncotarget.5683] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/13/2015] [Indexed: 12/23/2022] Open
Abstract
Background The relative contribution of isocitrate dehydrogenase mutations (mIDH) and O6-methylguanine-DNA methyltransferase promoter methylation (methMGMT) as biomarkers in glioblastoma remain poorly understood. Methods We investigated the association between methMGMT and mIDH with progression free survival and overall survival in a prospectively collected molecular registry of 274 glioblastoma patients. Results For glioblastoma patients who underwent Temozolomide and Radiation Therapy, OS and PFS was most favorable for those with tumors harboring both mIDH and methMGMT (median OS: 35.8 mo, median PFS: 27.5 mo); patients afflicted glioblastomas with either mIDH or methMGMT exhibited intermediate OS and PFS (mOS: 36 and 17.1 mo; mPFS: 12.2 mo and 9.9 mo, respectively); poorest OS and PFS was observed in wild type IDH1 (wtIDH1) glioblastomas that were MGMT promoter unmethylated (mOS: 15 mo, mPFS: 9.7 mo). For patients with wtIDH glioblastomas, TMZ+RT was associated with improved OS and PFS relative to patients treated with RT (OS: 15.4 mo v 9.6 mo, p < 0.001; PFS: 9.9 mo v 6.5 mo, p < 0.001). While TMZ+RT and RT treated mIDH patients exhibited improved overall survival relative to those with wtIDH, there were no differences between the TMZ+RT or RT group. These results suggest that mIDH1 conferred resistance to TMZ. Supporting this hypothesis, exogenous expression of mIDH1 in independent astrocytoma/glioblastoma lines resulted in a 3–10 fold increase in TMZ resistance after long-term passage. Conclusion Our study demonstrates IDH mutation and MGMT promoter methylation status independently associate with favorable outcome in TMZ+RT treated glioblastoma patients. However, these biomarkers differentially impact clinical TMZ response.
Collapse
Affiliation(s)
- Pei Yang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yinyan Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoxia Peng
- Department of Epidemiology and Biostatistics, School of Public Health and Family Medicine, Capital Medical University, Beijing, China
| | - Baoshi Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoguang Qiu
- Department of Radiation Therapy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guilin Li
- Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chenxing Wu
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Kun Yao
- Department of Pathology, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wei Yan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Li
- Center for Theoretic and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego, CA, USA
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Clark C Chen
- Center for Theoretic and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego, CA, USA
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
22
|
Ma QQ, Huang JT, Xiong YG, Yang XY, Han R, Zhu WW. MicroRNA-96 Regulates Apoptosis by Targeting PDCD4 in Human Glioma Cells. Technol Cancer Res Treat 2016; 16:92-98. [PMID: 26846266 DOI: 10.1177/1533034616629260] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Glioblastoma multiforme, the most common and aggressive form of primary brain tumor, presents a dismal prognosis. MicroRNAs play a critical role in the initiation, progression, and metastasis of cancer; however, the potential biological role of miRNAs in glioblastoma multiforme remains largely unknown. In our study, we found that microRNA-96 is upregulated in glioma tissues than in normal human brains. Transfection of microRNA-96 mimics into glioma cells significantly decreases apoptosis by suppressing PDCD4, a well-known tumor suppressor that is involved in apoptosis. In contrast, knockdown of microRNA-96 enhanced apoptosis. In vivo, microRNA-96 overexpression inhibits the apoptosis and increases tumor growth. These data suggest that microRNA-96 is a potential molecular target for glioma treatment.
Collapse
Affiliation(s)
- Qing-Qing Ma
- 1 Clinical Laboratory, Guizhou Aerospace Hospital, Zunyi, China
| | - Jian-Ting Huang
- 2 Department of Anesthesiology, The People's Hospital of Chengyang District, Qingdao, China
| | - Yun-Gang Xiong
- 1 Clinical Laboratory, Guizhou Aerospace Hospital, Zunyi, China
| | - Xiao-Yan Yang
- 1 Clinical Laboratory, Guizhou Aerospace Hospital, Zunyi, China
| | - Ran Han
- 1 Clinical Laboratory, Guizhou Aerospace Hospital, Zunyi, China
| | - Wang-Wen Zhu
- 1 Clinical Laboratory, Guizhou Aerospace Hospital, Zunyi, China
| |
Collapse
|
23
|
Abstract
All known cells continuously release nanoscale lipid membrane-enclosed packets. These packets, termed extracellular vesicles (EVs), bear the signature of their cells of origin. These vesicles can be detected in just about every type of biofluid tested, including blood, urine, and cerebrospinal fluid. The majority comes from normal cells, but disease cells also release them. There is a great interest in collecting and analyzing EVs in biofluids as diagnostics for a wide spectrum of central nervous system diseases. Here, we will review the state of central nervous system EV research in terms of molecular diagnostics and biomarkers.
Collapse
|
24
|
Hong L, Ya-wei L, Hai W, Qiang Z, Jun-jie L, Huang A, Song-tao Q, Yun-tao L. MiR-519a functions as a tumor suppressor in glioma by targeting the oncogenic STAT3 pathway. J Neurooncol 2016; 128:35-45. [DOI: 10.1007/s11060-016-2095-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/28/2016] [Indexed: 10/22/2022]
|
25
|
Hirshman BR, Tang JA, Jones LA, Proudfoot JA, Carley KM, Marshall L, Carter BS, Chen CC. Impact of medical academic genealogy on publication patterns: An analysis of the literature for surgical resection in brain tumor patients. Ann Neurol 2016; 79:169-77. [PMID: 26727354 DOI: 10.1002/ana.24569] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/07/2015] [Accepted: 11/15/2015] [Indexed: 11/09/2022]
Abstract
"Academic genealogy" refers to the linking of scientists and scholars based on their dissertation supervisors. We propose that this concept can be applied to medical training and that this "medical academic genealogy" may influence the landscape of the peer-reviewed literature. We performed a comprehensive PubMed search to identify US authors who have contributed peer-reviewed articles on a neurosurgery topic that remains controversial: the value of maximal resection for high-grade gliomas (HGGs). Training information for each key author (defined as the first or last author of an article) was collected (eg, author's medical school, residency, and fellowship training). Authors were recursively linked to faculty mentors to form genealogies. Correlations between genealogy and publication result were examined. Our search identified 108 articles with 160 unique key authors. Authors who were members of 2 genealogies (14% of key authors) contributed to 38% of all articles. If an article contained an authorship contribution from the first genealogy, its results were more likely to support maximal resection (log odds ratio = 2.74, p < 0.028) relative to articles without such contribution. In contrast, if an article contained an authorship contribution from the second genealogy, it was less likely to support maximal resection (log odds ratio = -1.74, p < 0.026). We conclude that the literature on surgical resection for HGGs is influenced by medical academic genealogies, and that articles contributed by authors of select genealogies share common results. These findings have important implications for the interpretation of scientific literature, design of medical training, and health care policy.
Collapse
Affiliation(s)
- Brian R Hirshman
- Center for Translational and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego, San Diego, CA.,Center for Computational Analysis of Social and Organizational Systems, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA.,Computation, Organizations, and Society Program, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA
| | - Jessica A Tang
- Center for Translational and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego, San Diego, CA
| | - Laurie A Jones
- Computation, Organizations, and Society Program, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA
| | - James A Proudfoot
- Clinical and Translational Research Institute, University of California, San Diego, San Diego, CA
| | - Kathleen M Carley
- Center for Computational Analysis of Social and Organizational Systems, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA.,Computation, Organizations, and Society Program, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA
| | - Lawrence Marshall
- Department of Neurosurgery, University of California, San Diego, San Diego, CA
| | - Bob S Carter
- Center for Translational and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego, San Diego, CA.,Department of Neurosurgery, University of California, San Diego, San Diego, CA
| | - Clark C Chen
- Center for Translational and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego, San Diego, CA.,Department of Neurosurgery, University of California, San Diego, San Diego, CA
| |
Collapse
|
26
|
Kim DG, Jin Y, Jin J, Yang H, Joo KM, Lee WS, Shim SR, Kim SW, Yoo J, Lee SH, Yoo JS, Nam DH. Anticancer activity of TTAC-0001, a fully human anti-vascular endothelial growth factor receptor 2 (VEGFR-2/KDR) monoclonal antibody, is associated with inhibition of tumor angiogenesis. MAbs 2015; 7:1195-204. [PMID: 26325365 PMCID: PMC4966428 DOI: 10.1080/19420862.2015.1086854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) and its receptors are considered the primary cause of tumor-induced angiogenesis. Specifically, VEGFR-2/kinase insert domain receptor (KDR) is part of the major signaling pathway that plays a significant role in tumor angiogenesis, which is associated with the development of various types of tumor and metastasis. In particular, KDR is involved in tumor angiogenesis as well as cancer cell growth and survival. In this study, we evaluated the therapeutic potential of TTAC-0001, a fully human antibody against VEGFR-2/KDR. To assess the efficacy of the antibody and pharmacokinetic (PK) relationship in vivo, we tested the potency of TTAC-0001 in glioblastoma and colorectal cancer xenograft models. Antitumor activity of TTAC-0001 in preclinical models correlated with tumor growth arrest, induction of tumor cell apoptosis, and inhibition of angiogenesis. We also evaluated the combination effect of TTAC-0001 with a chemotherapeutic agent in xenograft models. We were able to determine the relationship between PK and the efficacy of TTAC-0001 through in vivo single-dose PK study. Taken together, our data suggest that targeting VEGFR-2 with TTAC-0001 could be a promising approach for cancer treatment.
Collapse
Affiliation(s)
- Dong Geon Kim
- a Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center ; Seoul 06351 , Korea.,b Department of Health Sciences and Technology ; SAIHST, Sungkyunkwan University ; Seoul 06351 , Korea
| | - Younggeon Jin
- a Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center ; Seoul 06351 , Korea
| | - Juyoun Jin
- a Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center ; Seoul 06351 , Korea
| | - Heekyoung Yang
- a Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center ; Seoul 06351 , Korea.,c Department of Neurosurgery ; Samsung Medical Center, Sungkyunkwan University School of Medicine ; Seoul 06351 , Korea
| | - Kyeung Min Joo
- a Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center ; Seoul 06351 , Korea.,b Department of Health Sciences and Technology ; SAIHST, Sungkyunkwan University ; Seoul 06351 , Korea.,d Department of Anatomy and Cell Biology ; Sungkyunkwan University School of Medicine ; Seoul 06351 , Korea
| | - Weon Sup Lee
- e PharmAbcine; Inc., Daejeon Bioventure Town ; Daejeon 34054 , Korea
| | - Sang Ryeol Shim
- e PharmAbcine; Inc., Daejeon Bioventure Town ; Daejeon 34054 , Korea
| | - Sung-Woo Kim
- e PharmAbcine; Inc., Daejeon Bioventure Town ; Daejeon 34054 , Korea
| | - Jinsang Yoo
- e PharmAbcine; Inc., Daejeon Bioventure Town ; Daejeon 34054 , Korea
| | - Sang Hoon Lee
- f Bio Division, Hanwha Chemical ; Seoul 04541 , Korea
| | - Jin-San Yoo
- e PharmAbcine; Inc., Daejeon Bioventure Town ; Daejeon 34054 , Korea
| | - Do-Hyun Nam
- a Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center ; Seoul 06351 , Korea.,b Department of Health Sciences and Technology ; SAIHST, Sungkyunkwan University ; Seoul 06351 , Korea.,c Department of Neurosurgery ; Samsung Medical Center, Sungkyunkwan University School of Medicine ; Seoul 06351 , Korea
| |
Collapse
|
27
|
Li J, Taich ZJ, Goyal A, Gonda D, Akers J, Adhikari B, Patel K, Vandenberg S, Yan W, Bao Z, Carter BS, Wang R, Mao Y, Jiang T, Chen CC. Epigenetic suppression of EGFR signaling in G-CIMP+ glioblastomas. Oncotarget 2015; 5:7342-56. [PMID: 25277177 PMCID: PMC4202127 DOI: 10.18632/oncotarget.2350] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The intrinsic signaling cascades and cell states associated with the Glioma CpG Island Methylator Phenotype (G-CIMP) remain poorly understood. Using published mRNA signatures associated with EGFR activation, we demonstrate that G-CIMP+ tumors harbor decreased EGFR signaling using three independent datasets, including the Chinese Glioma Genome Atlas(CGGA; n=155), the REMBRANDT dataset (n=288), and The Cancer Genome Atlas (TCGA; n=406). Additionally, an independent collection of 25 fresh-frozen glioblastomas confirmed lowered pERK levels in G-CIMP+ specimens (p<0.001), indicating suppressed EGFR signaling. Analysis of TCGA glioblastomas revealed that G-CIMP+ glioblastomas harbored lowered mRNA levels for EGFR and H-Ras. Induction of G-CIMP+ state by exogenous expression of a mutated isocitrate dehydrogenase 1, IDH1-R132H, suppressed EGFR and H-Ras protein expression as well as pERK accumulation in independent glioblastoma models. These suppressions were associated with increased deposition of the repressive histone markers, H3K9me3 and H3K27me3, in the EGFR and H-Ras promoter regions. The IDH1-R132H expression-induced pERK suppression can be reversed by exogenous expression of H-RasG12V. Finally, the G-CIMP+ Ink4a-Arf−/− EGFRvIII glioblastoma line was more resistant to the EGFR inhibitor, Gefitinib, relative to its isogenic G-CIMP- counterpart. These results suggest that G-CIMP epigenetically regulates EGFR signaling and serves as a predictive biomarker for EGFR inhibitors in glioblastoma patients.
Collapse
Affiliation(s)
- Jie Li
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego. Contributed equally to this work
| | - Zachary J Taich
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego. Contributed equally to this work
| | - Amit Goyal
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego
| | - David Gonda
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego
| | - Johnny Akers
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego
| | - Bandita Adhikari
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego
| | - Kunal Patel
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego
| | | | - Wei Yan
- Department of Neurosurgery, Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhaoshi Bao
- Department of Neurosurgery, Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bob S Carter
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Beijing, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tao Jiang
- Department of Neurosurgery, Tiantan Hospital, Capital Medical University, Beijing, China
| | - Clark C Chen
- Center for Theoretical and Applied Neuro-Oncology, Division of Neurosurgery, University of California, San Diego
| |
Collapse
|
28
|
Myung K. Targeting the cancer cell state. Cell Cycle 2015; 14:2385-6. [PMID: 26083258 DOI: 10.1080/15384101.2015.1063294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Kyungjae Myung
- a Center for Genomic Integrity ; Institute for Basic Science ; Ulsan , Korea
| |
Collapse
|
29
|
Dynamic epigenetic regulation of glioblastoma tumorigenicity through LSD1 modulation of MYC expression. Proc Natl Acad Sci U S A 2015; 112:E4055-64. [PMID: 26159421 DOI: 10.1073/pnas.1501967112] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The available evidence suggests that the lethality of glioblastoma is driven by small subpopulations of cells that self-renew and exhibit tumorigenicity. It remains unclear whether tumorigenicity exists as a static property of a few cells or as a dynamically acquired property. We used tumor-sphere and xenograft formation as assays for tumorigenicity and examined subclones isolated from established and primary glioblastoma lines. Our results indicate that glioblastoma tumorigenicity is largely deterministic, yet the property can be acquired spontaneously at low frequencies. Further, these dynamic transitions are governed by epigenetic reprogramming through the lysine-specific demethylase 1 (LSD1). LSD depletion increases trimethylation of histone 3 lysine 4 at the avian myelocytomatosis viral oncogene homolog (MYC) locus, which elevates MYC expression. MYC, in turn, regulates oligodendrocyte lineage transcription factor 2 (OLIG2), SRY (sex determining region Y)-box 2 (SOX2), and POU class 3 homeobox 2 (POU3F2), a core set of transcription factors required for reprogramming glioblastoma cells into stem-like states. Our model suggests epigenetic regulation of key transcription factors governs transitions between tumorigenic states and provides a framework for glioblastoma therapeutic development.
Collapse
|
30
|
Dong X, Noorbakhsh A, Hirshman BR, Zhou T, Tang JA, Chang DC, Carter BS, Chen CC. Survival trends of grade I, II, and III astrocytoma patients and associated clinical practice patterns between 1999 and 2010: A SEER-based analysis. Neurooncol Pract 2015; 3:29-38. [PMID: 31579519 DOI: 10.1093/nop/npv016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Indexed: 11/13/2022] Open
Abstract
Background The survival trends and the patterns of clinical practice pertaining to radiation therapy and surgical resection for WHO grade I, II, and III astrocytoma patients remain poorly characterized. Methods Using the Surveillance, Epidemiology and End Results (SEER) database, we identified 2497 grade I, 4113 grade II, and 2755 grade III astrocytomas during the period of 1999-2010. Time-trend analyses were performed for overall survival, radiation treatment (RT), and the extent of surgical resection (EOR). Results While overall survival of grade I astrocytoma patients remained unchanged during the study period, we observed improved overall survival for grade II and III astrocytoma patients (Tarone-Ware P < .05). The median survival increased from 44 to 57 months and from 15 to 24 months for grade II and III astrocytoma patients, respectively. The differences in survival remained significant after adjusting for pertinent variables including age, ethnicity, marital status, sex, tumor size, tumor location, EOR, and RT status. The pattern of clinical practice in terms of EOR for grade II and III astrocytoma patients did not change significantly during this study period. However, there was decreased RT utilization as treatment for grade II astrocytoma patients after 2005. Conclusion Results from the SEER database indicate that there were improvements in the overall survival of grade II and III astrocytoma patients over the past decade. Analysis of the clinical practice patterns identified potential opportunities for impacting the clinical course of these patients.
Collapse
Affiliation(s)
- Xuezhi Dong
- School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093 (X.D., A.N., B.R.H., T.Z., J.A.T.); Division of Neurological Surgery, University of California, San Diego, 200 West Arbor Drive #8893, San Diego, California 92103 (B.S.C., C.C.C.); Department of Surgery, University of California, San Diego, 200 West Arbor Drive #8220, San Diego, California 92103 (D.C.C.)
| | - Abraham Noorbakhsh
- School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093 (X.D., A.N., B.R.H., T.Z., J.A.T.); Division of Neurological Surgery, University of California, San Diego, 200 West Arbor Drive #8893, San Diego, California 92103 (B.S.C., C.C.C.); Department of Surgery, University of California, San Diego, 200 West Arbor Drive #8220, San Diego, California 92103 (D.C.C.)
| | - Brian R Hirshman
- School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093 (X.D., A.N., B.R.H., T.Z., J.A.T.); Division of Neurological Surgery, University of California, San Diego, 200 West Arbor Drive #8893, San Diego, California 92103 (B.S.C., C.C.C.); Department of Surgery, University of California, San Diego, 200 West Arbor Drive #8220, San Diego, California 92103 (D.C.C.)
| | - Tianzan Zhou
- School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093 (X.D., A.N., B.R.H., T.Z., J.A.T.); Division of Neurological Surgery, University of California, San Diego, 200 West Arbor Drive #8893, San Diego, California 92103 (B.S.C., C.C.C.); Department of Surgery, University of California, San Diego, 200 West Arbor Drive #8220, San Diego, California 92103 (D.C.C.)
| | - Jessica A Tang
- School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093 (X.D., A.N., B.R.H., T.Z., J.A.T.); Division of Neurological Surgery, University of California, San Diego, 200 West Arbor Drive #8893, San Diego, California 92103 (B.S.C., C.C.C.); Department of Surgery, University of California, San Diego, 200 West Arbor Drive #8220, San Diego, California 92103 (D.C.C.)
| | - David C Chang
- School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093 (X.D., A.N., B.R.H., T.Z., J.A.T.); Division of Neurological Surgery, University of California, San Diego, 200 West Arbor Drive #8893, San Diego, California 92103 (B.S.C., C.C.C.); Department of Surgery, University of California, San Diego, 200 West Arbor Drive #8220, San Diego, California 92103 (D.C.C.)
| | - Bob S Carter
- School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093 (X.D., A.N., B.R.H., T.Z., J.A.T.); Division of Neurological Surgery, University of California, San Diego, 200 West Arbor Drive #8893, San Diego, California 92103 (B.S.C., C.C.C.); Department of Surgery, University of California, San Diego, 200 West Arbor Drive #8220, San Diego, California 92103 (D.C.C.)
| | - Clark C Chen
- School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093 (X.D., A.N., B.R.H., T.Z., J.A.T.); Division of Neurological Surgery, University of California, San Diego, 200 West Arbor Drive #8893, San Diego, California 92103 (B.S.C., C.C.C.); Department of Surgery, University of California, San Diego, 200 West Arbor Drive #8220, San Diego, California 92103 (D.C.C.)
| |
Collapse
|
31
|
Siu A, Volotskova O, Cheng X, Khalsa SS, Bian K, Murad F, Keidar M, Sherman JH. Differential Effects of Cold Atmospheric Plasma in the Treatment of Malignant Glioma. PLoS One 2015; 10:e0126313. [PMID: 26083405 PMCID: PMC4471333 DOI: 10.1371/journal.pone.0126313] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/31/2015] [Indexed: 11/19/2022] Open
Abstract
Objective Cold atmospheric plasma (CAP) has recently been shown to selectively target cancer cells with minimal effects on normal cells. We systematically assessed the effects of CAP in the treatment of glioblastoma. Methods Three glioma cell lines, normal astrocytes, and endothelial cell lines were treated with CAP. The effects of CAP were then characterized for viability, cytotoxicity/apoptosis, and cell cycle effects. Statistical significance was determined with student's t-test. Results CAP treatment decreases viability of glioma cells in a dose dependent manner, with the ID50 between 90-120 seconds for all glioma cell lines. Treatment with CAP for more than 120 seconds resulted in viability less than 35% at 24-hours posttreatment, with a steady decline to less than 20% at 72-hours. In contrast, the effect of CAP on the viability of NHA and HUVEC was minimal, and importantly not significant at 90 to 120 seconds, with up to 85% of the cells remained viable at 72-hours post-treatment. CAP treatment produces both cytotoxic and apoptotic effects with some variability between cell lines. CAP treatment resulted in a G2/M-phase cell cycle pause in all three cell lines. Conclusions This preliminary study determined a multi-focal effect of CAP on glioma cells in vitro, which was not observed in the non-tumor cell lines. The decreased viability depended on the treatment duration and cell line, but overall was explained by the induction of cytotoxicity, apoptosis, and G2/M pause. Future studies will aim at further characterization with more complex pre-clinical models.
Collapse
Affiliation(s)
- Alan Siu
- Department of Neurological Surgery, George Washington University, Washington, DC, United States of America
| | - Olga Volotskova
- Department of Mechanical and Aerospace Engineering, George Washington University, Washington, DC, United States of America
| | - Xiaoqian Cheng
- Department of Mechanical and Aerospace Engineering, George Washington University, Washington, DC, United States of America
| | - Siri S. Khalsa
- George Washington University School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Ka Bian
- Department of Biochemistry and Molecular Biology, George Washington University, School of Medicine, Washington, DC, United States of America
| | - Ferid Murad
- Department of Biochemistry and Molecular Biology, George Washington University, School of Medicine, Washington, DC, United States of America
| | - Michael Keidar
- Department of Mechanical and Aerospace Engineering, George Washington University, Washington, DC, United States of America
| | - Jonathan H. Sherman
- Department of Neurological Surgery, George Washington University, Washington, DC, United States of America
- * E-mail:
| |
Collapse
|
32
|
Shen Y, Li J, Nitta M, Futalan D, Steed T, Treiber JM, Taich Z, Stevens D, Wykosky J, Chen HZ, Carter BS, Becher OJ, Kennedy R, Esashi F, Sarkaria JN, Furnari FB, Cavenee WK, Desai A, Chen CC. Orthogonal targeting of EGFRvIII expressing glioblastomas through simultaneous EGFR and PLK1 inhibition. Oncotarget 2015; 6:11751-67. [PMID: 26059434 PMCID: PMC4494902 DOI: 10.18632/oncotarget.3996] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 04/20/2015] [Indexed: 11/25/2022] Open
Abstract
We identified a synthetic lethality between PLK1 silencing and the expression of an oncogenic Epidermal Growth Factor Receptor, EGFRvIII. PLK1 promoted homologous recombination (HR), mitigating EGFRvIII induced oncogenic stress resulting from DNA damage accumulation. Accordingly, PLK1 inhibition enhanced the cytotoxic effects of the DNA damaging agent, temozolomide (TMZ). This effect was significantly more pronounced in an Ink4a/Arf(-/-) EGFRvIII glioblastoma model relative to an Ink4a/Arf(-/-) PDGF-β model. The tumoricidal and TMZ-sensitizing effects of BI2536 were uniformly observed across Ink4a/Arf(-/-) EGFRvIII glioblastoma clones that acquired independent resistance mechanisms to EGFR inhibitors, suggesting these resistant clones retain oncogenic stress that required PLK1 compensation. Although BI2536 significantly augmented the anti-neoplastic effect of EGFR inhibitors in the Ink4a/Arf(-/-) EGFRvIII model, durable response was not achieved until TMZ was added. Our results suggest that optimal therapeutic effect against glioblastomas requires a "multi-orthogonal" combination tailored to the molecular physiology associated with the target cancer genome.
Collapse
Affiliation(s)
- Ying Shen
- Center for Theoretical and Applied Neuro-Oncology, Moores Cancer Center, Division of Neurosurgery, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Li
- Center for Theoretical and Applied Neuro-Oncology, Moores Cancer Center, Division of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| | - Masayuki Nitta
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Diahnn Futalan
- Center for Theoretical and Applied Neuro-Oncology, Moores Cancer Center, Division of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| | - Tyler Steed
- Center for Theoretical and Applied Neuro-Oncology, Moores Cancer Center, Division of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| | - Jeffrey M. Treiber
- Center for Theoretical and Applied Neuro-Oncology, Moores Cancer Center, Division of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| | - Zack Taich
- Center for Theoretical and Applied Neuro-Oncology, Moores Cancer Center, Division of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| | - Deanna Stevens
- San Diego Branch, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Jill Wykosky
- San Diego Branch, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bob S. Carter
- Center for Theoretical and Applied Neuro-Oncology, Moores Cancer Center, Division of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| | - Oren J. Becher
- Departments of Pediatrics and Pathology, Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
| | - Richard Kennedy
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - Fumiko Esashi
- The Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Frank B. Furnari
- San Diego Branch, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Webster K. Cavenee
- San Diego Branch, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Arshad Desai
- San Diego Branch, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Clark C. Chen
- Center for Theoretical and Applied Neuro-Oncology, Moores Cancer Center, Division of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
33
|
Cheng W, Li M, Cai J, Wang K, Zhang C, Bao Z, Liu Y, Wu A. HDAC4, a prognostic and chromosomal instability marker, refines the predictive value of MGMT promoter methylation. J Neurooncol 2015; 122:303-12. [PMID: 25557107 PMCID: PMC4368847 DOI: 10.1007/s11060-014-1709-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 12/24/2014] [Indexed: 01/23/2023]
Abstract
Chromosomal instability is a hallmark of human cancers and is closely linked to tumorigenesis. The prognostic value of molecular signatures of chromosomal instability (CIN) has been validated in various cancers. However, few studies have examined the relationship between CIN and glioma. Histone deacetylases (HDACs) regulate chromosome structure and are linked to the loss of genomic integrity in cancer cells. In this study, the prognostic value of HDAC4 expression and its association with markers of CIN were investigated by analyzing data from our own and four other large sample databases. The results showed that HDAC4 expression is downregulated in high- as compared to low-grade glioma and is associated with a favorable clinical outcome. HDAC4 expression and CIN were closely related in glioma from both functional and statistical standpoints. Moreover, the predictive value of the O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status-a widely used glioma marker-was refined by HDAC4 expression level, which was significantly related to CIN in our study. In conclusion, we propose that HDAC4 expression, a prognostic and CIN marker, enhances the predictive value of MGMT promoter methylation status for identifying patients who will most benefit from radiochemotherapy.
Collapse
Affiliation(s)
- Wen Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001 China
- Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Mingyang Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Kuanyu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Chuanbao Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Zhaoshi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Yanwei Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Anhua Wu
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001 China
- Chinese Glioma Cooperative Group (CGCG), Beijing, China
| |
Collapse
|
34
|
Li J, Zhu S, Kozono D, Ng K, Futalan D, Shen Y, Akers JC, Steed T, Kushwaha D, Schlabach M, Carter BS, Kwon CH, Furnari F, Cavenee W, Elledge S, Chen CC. Genome-wide shRNA screen revealed integrated mitogenic signaling between dopamine receptor D2 (DRD2) and epidermal growth factor receptor (EGFR) in glioblastoma. Oncotarget 2015; 5:882-93. [PMID: 24658464 PMCID: PMC4011590 DOI: 10.18632/oncotarget.1801] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma remains one of the deadliest of human cancers, with most patients succumbing to the disease within two years of diagnosis. The available data suggest that simultaneous inactivation of critical nodes within the glioblastoma molecular circuitry will be required for meaningful clinical efficacy. We conducted parallel genome-wide shRNA screens to identify such nodes and uncovered a number of G-Protein Coupled Receptor (GPCR) neurotransmitter pathways, including the Dopamine Receptor D2 (DRD2) signaling pathway. Supporting the importance of DRD2 in glioblastoma, DRD2 mRNA and protein expression were elevated in clinical glioblastoma specimens relative to matched non-neoplastic cerebrum. Treatment with independent si-/shRNAs against DRD2 or with DRD2 antagonists suppressed the growth of patient-derived glioblastoma lines both in vitro and in vivo. Importantly, glioblastoma lines derived from independent genetically engineered mouse models (GEMMs) were more sensitive to haloperidol, an FDA approved DRD2 antagonist, than the premalignant astrocyte lines by approximately an order of magnitude. The pro-proliferative effect of DRD2 was, in part, mediated through a GNAI2/Rap1/Ras/ERK signaling axis. Combined inhibition of DRD2 and Epidermal Growth Factor Receptor (EGFR) led to synergistic tumoricidal activity as well as ERK suppression in independent in vivo and in vitro glioblastoma models. Our results suggest combined EGFR and DRD2 inhibition as a promising strategy for glioblastoma treatment.
Collapse
|
35
|
Gray GK, McFarland BC, Nozell SE, Benveniste EN. NF-κB and STAT3 in glioblastoma: therapeutic targets coming of age. Expert Rev Neurother 2014; 14:1293-306. [PMID: 25262780 DOI: 10.1586/14737175.2014.964211] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since we last addressed the roles of NF-κB and JAK/STAT3 signaling in glioblastoma (GBM) 5 years ago, tremendous strides have been made in the understanding of these two pathways in glioma biology. Contributing to prosurvival mechanisms, cancer stem cell maintenance and treatment resistance, both NF-κB and STAT3 have been characterized as major drivers of GBM. In this review, we address general improvements in the molecular understanding of GBM, the structure of NF-κB and STAT3 signaling, the ways in which these pathways contribute to GBM and advances in preclinical and clinical targeting of these two signaling cascades.
Collapse
Affiliation(s)
- G Kenneth Gray
- Department of Cell, Developmental and Integrative Biology, 1900 University Blvd, THT 926A, University of Alabama at Birmingham, Birmingham, AL, 35294-0006, USA
| | | | | | | |
Collapse
|
36
|
Bao ZS, Chen HM, Yang MY, Zhang CB, Yu K, Ye WL, Hu BQ, Yan W, Zhang W, Akers J, Ramakrishnan V, Li J, Carter B, Liu YW, Hu HM, Wang Z, Li MY, Yao K, Qiu XG, Kang CS, You YP, Fan XL, Song WS, Li RQ, Su XD, Chen CC, Jiang T. RNA-seq of 272 gliomas revealed a novel, recurrent PTPRZ1-MET fusion transcript in secondary glioblastomas. Genome Res 2014; 24:1765-73. [PMID: 25135958 PMCID: PMC4216918 DOI: 10.1101/gr.165126.113] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Studies of gene rearrangements and the consequent oncogenic fusion proteins have laid the foundation for targeted cancer therapy. To identify oncogenic fusions associated with glioma progression, we catalogued fusion transcripts by RNA-seq of 272 gliomas. Fusion transcripts were more frequently found in high-grade gliomas, in the classical subtype of gliomas, and in gliomas treated with radiation/temozolomide. Sixty-seven in-frame fusion transcripts were identified, including three recurrent fusion transcripts: FGFR3-TACC3, RNF213-SLC26A11, and PTPRZ1-MET (ZM). Interestingly, the ZM fusion was found only in grade III astrocytomas (1/13; 7.7%) or secondary GBMs (sGBMs, 3/20; 15.0%). In an independent cohort of sGBMs, the ZM fusion was found in three of 20 (15%) specimens. Genomic analysis revealed that the fusion arose from translocation events involving introns 3 or 8 of PTPRZ and intron 1 of MET. ZM fusion transcripts were found in GBMs irrespective of isocitrate dehydrogenase 1 (IDH1) mutation status. sGBMs harboring ZM fusion showed higher expression of genes required for PIK3CA signaling and lowered expression of genes that suppressed RB1 or TP53 function. Expression of the ZM fusion was mutually exclusive with EGFR overexpression in sGBMs. Exogenous expression of the ZM fusion in the U87MG glioblastoma line enhanced cell migration and invasion. Clinically, patients afflicted with ZM fusion harboring glioblastomas survived poorly relative to those afflicted with non-ZM-harboring sGBMs (P < 0.001). Our study profiles the shifting RNA landscape of gliomas during progression and reveled ZM as a novel, recurrent fusion transcript in sGBMs.
Collapse
Affiliation(s)
- Zhao-Shi Bao
- Beijing Neurosurgical Institute, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Hui-Min Chen
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China;
| | - Ming-Yu Yang
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Chuan-Bao Zhang
- Beijing Neurosurgical Institute, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Kai Yu
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Wan-Lu Ye
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Bo-Qiang Hu
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Wei Yan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Johnny Akers
- Center for Theoretical and Applied Neuro-Oncology (CTAN), Division of Neurosurgery, University of California, San Diego, California 92093, USA
| | - Valya Ramakrishnan
- Center for Theoretical and Applied Neuro-Oncology (CTAN), Division of Neurosurgery, University of California, San Diego, California 92093, USA
| | - Jie Li
- Center for Theoretical and Applied Neuro-Oncology (CTAN), Division of Neurosurgery, University of California, San Diego, California 92093, USA
| | - Bob Carter
- Center for Theoretical and Applied Neuro-Oncology (CTAN), Division of Neurosurgery, University of California, San Diego, California 92093, USA
| | - Yan-Wei Liu
- Beijing Neurosurgical Institute, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Hui-Min Hu
- Beijing Neurosurgical Institute, Beijing 100050, China
| | - Zheng Wang
- Beijing Neurosurgical Institute, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Ming-Yang Li
- Beijing Neurosurgical Institute, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Kun Yao
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China; Department of Pathology, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Xiao-Guang Qiu
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China; Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Chun-Sheng Kang
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - Yong-Ping You
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Long Fan
- Laboratory of Neuroscience and Brain Development, Beijing Key Laboratory of Gene Resources and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Wei Sonya Song
- Beijing Neurosurgical Institute, Beijing 100050, China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Rui-Qiang Li
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Xiao-Dong Su
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China;
| | - Clark C Chen
- Center for Theoretical and Applied Neuro-Oncology (CTAN), Division of Neurosurgery, University of California, San Diego, California 92093, USA;
| | - Tao Jiang
- Beijing Neurosurgical Institute, Beijing 100050, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing 100069, China; China National Clinical Research Center for Neurological Diseases, Beijing 100050, China
| |
Collapse
|
37
|
Kaye AH, Morokoff A. The Continuing Evolution: Biology and Treatment of Brain Tumors. Neurosurgery 2014; 61 Suppl 1:100-4. [DOI: 10.1227/neu.0000000000000388] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Andrew H. Kaye
- Department of Neurosurgery, Royal Melbourne Hospital, Parkville, Australia
| | - Andrew Morokoff
- Department of Surgery, University of Melbourne, Melbourne, Australia
| |
Collapse
|
38
|
Abstract
BACKGROUND Glioblastoma is the most common primary brain tumour. It has a poor prognosis despite some advances in treatment that have been achieved over the last ten years. In Slovenia, 50 to 60 glioblastoma patients are diagnosed each year. In order to establish whether the current treatment options have any influence on the survival of the Slovenian glioblastoma patients, their data in the period from the beginning of the year 1997 to the end of the year 2008 have been analysed. PATIENTS AND METHODS All patients treated at the Institute of Oncology Ljubljana from 1997 to 2008 were included in the retrospective study. Demographics, treatment details, and survival time after the diagnosis were collected and statistically analysed for the group as a whole and for subgroups. RESULTS From 1997 to 2008, 527 adult patients were diagnosed with glioblastoma and referred to the Institute of Oncology for further treatment. Their median age was 59 years (from 20 to 85) and all but one had the diagnosis confirmed by a pathologist. Gross total resection was reported by surgeons in 261 (49.5%) patients; good functional status (WHO 0 or 1) after surgery was observed in 336 (63.7%) patients, radiotherapy was performed in 422 (80.1%) patients, in 317 (75.1%) of them with radical intent, and 198 (62.5 %) of those received some form of systemic treatment (usually temozolomide). The median survival of all patients amounted to 9.7 months. There was no difference in median survival of all patients or of all treated patients before or after the chemo-radiotherapy era. However, the overall survival of patients treated with radical intent was significantly better (11.4 months; p < 0.05). A better survival was also noticed in radically treated patients who received additional temozolomide therapy (11.4 vs. 13.1 months; p = 0.014). The longer survival was associated with a younger age and a good performance status as well as with a more extensive tumour resection. In patients treated with radical intent, having a good performance status, and receiving radiotherapy and additional temozolomide therapy, the survival was significantly longer, based on multivariate analysis. CONCLUSIONS We observed a gradual increase in the survival of glioblastoma patients who were treated with radical intent over the last ten years. Good functional surgery, advances in radiotherapy and addition of temozolomide all contributed to this increase. Though the increased survival seems to be more pronounced in certain subgroups, we have still not been able to exactly define them. Further research, especially in tumour biology and genetics is needed.
Collapse
|
39
|
Milinkovic V, Bankovic J, Rakic M, Stankovic T, Skender-Gazibara M, Ruzdijic S, Tanic N. Identification of novel genetic alterations in samples of malignant glioma patients. PLoS One 2013; 8:e82108. [PMID: 24358143 PMCID: PMC3864906 DOI: 10.1371/journal.pone.0082108] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 10/25/2013] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma is the most frequent and malignant human brain tumor. High level of genomic instability detected in glioma cells implies that numerous genetic alterations accumulate during glioma pathogenesis. We investigated alterations in AP-PCR DNA profiles of 30 glioma patients, and detected specific changes in 11 genes not previously associated with this disease: LHFPL3, SGCG, HTR4, ITGB1, CPS1, PROS1, GP2, KCNG2, PDE4D, KIR3DL3, and INPP5A. Further correlations revealed that 8 genes might play important role in pathogenesis of glial tumors, while changes in GP2, KCNG2 and KIR3DL3 should be considered as passenger mutations, consequence of high level of genomic instability. Identified genes have a significant role in signal transduction or cell adhesion, which are important processes for cancer development and progression. According to our results, LHFPL3 might be characteristic of primary glioblastoma, SGCG, HTR4, ITGB1, CPS1, PROS1 and INPP5A were detected predominantly in anaplastic astrocytoma, suggesting their role in progression of secondary glioblastoma, while alterations of PDE4D seem to have important role in development of both glioblastoma subtypes. Some of the identified genes showed significant association with p53, p16, and EGFR, but there was no significant correlation between loss of PTEN and any of identified genes. In conclusion our study revealed genetic alterations that were not previously associated with glioma pathogenesis and could be potentially used as molecular markers of different glioblastoma subtypes.
Collapse
Affiliation(s)
- Vedrana Milinkovic
- University of Belgrade, Institute for Biological Research “Sinisa Stankovic”, Department of Neurobiology, Belgrade, Republic of Serbia
| | - Jasna Bankovic
- University of Belgrade, Institute for Biological Research “Sinisa Stankovic”, Department of Neurobiology, Belgrade, Republic of Serbia
| | - Miodrag Rakic
- Clinical Center of Serbia, Clinic for Neurosurgery, Belgrade, Republic of Serbia
| | - Tijana Stankovic
- University of Belgrade, Institute for Biological Research “Sinisa Stankovic”, Department of Neurobiology, Belgrade, Republic of Serbia
| | - Milica Skender-Gazibara
- University of Belgrade, School of Medicine, Institute of Pathology, Belgrade, Republic of Serbia
| | - Sabera Ruzdijic
- University of Belgrade, Institute for Biological Research “Sinisa Stankovic”, Department of Neurobiology, Belgrade, Republic of Serbia
| | - Nikola Tanic
- University of Belgrade, Institute for Biological Research “Sinisa Stankovic”, Department of Neurobiology, Belgrade, Republic of Serbia
| |
Collapse
|