1
|
Zhou K, Wan J, Li Y, Yuan Y, Liu Q, Li H, Jiang X, Yuan X, Zhang S, Zhang Y. Prognostic value of pre-treatment neutrophil-to-lymphocyte ratio in patients with brain metastasis from cancer: a meta-analysis. Sci Rep 2024; 14:24789. [PMID: 39433560 PMCID: PMC11494139 DOI: 10.1038/s41598-024-76305-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
Evidence shows that inflammatory responses play an essential role in the development of brain metastases (BM). The goal of this meta-analysis was to critically evaluate the literature regarding the use of the neutrophil to lymphocyte ratio (NLR) to predict the prognosis of patients with BM to help clinicians institute early interventions and improve outcomes. We conducted systematic review and meta-analysis, utilizing data from prominent databases, including PubMed, Cochrane Library and China National Knowledge Infrastructure databases. Our inclusion criteria encompassed studies investigating the studies that assessed the association between NLR and overall survival (OS). We included 11 articles, with 2629 eligible patients, to evaluate the association between NLR and OS. High NLR was significantly associated with shorter OS, with a pooled hazard ratio (HR) of 1.82 (95% CI 1.57-2.11). Subgroup analysis revealed that this association was consistent across different regions, with HRs of 2.03 (95% CI 1.67-2.46) in Asian populations and 1.58 (95% CI 1.35-1.84) in non-Asian populations. Additionally, in a subgroup analysis based on NLR cut-off values, patients with NLR ≥ 3 had an HR of 1.69 (95% CI 1.46-1.96), while those with NLR < 3 had an HR of 2.26 (95% CI 1.64-3.11). Sensitivity analysis confirmed that no single study significantly influenced the pooled effect size. Our meta-analysis confirmed the prognostic value of NLR in patients with brain metastasis.
Collapse
Affiliation(s)
- Ke Zhou
- Center for Evidence-Based Medicine, Clinical Research Center, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
- School of Preclinical Medicine / School of Nursing, Chengdu University, Chengdu, Sichuan, China
| | - Jun Wan
- Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Yile Li
- School of Preclinical Medicine / School of Nursing, Chengdu University, Chengdu, Sichuan, China
| | - Yi Yuan
- School of Preclinical Medicine / School of Nursing, Chengdu University, Chengdu, Sichuan, China
| | - Qian Liu
- Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Huixuan Li
- Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Xinyi Jiang
- Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Xiang Yuan
- Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Sen Zhang
- Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Yu Zhang
- Center for Evidence-Based Medicine, Clinical Research Center, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Morrison C, Weterings E, Gravbrot N, Hammer M, Weinand M, Sanan A, Pandey R, Mahadevan D, Stea B. Gene Expression Patterns Associated with Survival in Glioblastoma. Int J Mol Sci 2024; 25:3668. [PMID: 38612480 PMCID: PMC11011684 DOI: 10.3390/ijms25073668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
The aim of this study was to investigate gene expression alterations associated with overall survival (OS) in glioblastoma (GBM). Using the Nanostring nCounter platform, we identified four genes (COL1A2, IGFBP3, NGFR, and WIF1) that achieved statistical significance when comparing GBM with non-neoplastic brain tissue. The four genes were included in a multivariate Cox Proportional Hazard model, along with age, extent of resection, and O6-methylguanine-DNA methyltransferase (MGMT) promotor methylation, to create a unique glioblastoma prognostic index (GPI). The GPI score inversely correlated with survival: patient with a high GPI had a median OS of 7.5 months (18-month OS = 9.7%) whereas patients with a low GPI had a median OS of 20.1 months (18-month OS = 54.5%; log rank p-value = 0.004). The GPI score was then validated in 188 GBM patients from The Cancer Genome Atlas (TCGA) from a national data base; similarly, patients with a high GPI had a median OS of 10.5 months (18-month OS = 12.4%) versus 16.9 months (18-month OS = 41.5%) for low GPI (log rank p-value = 0.0003). We conclude that this novel mRNA-based prognostic index could be useful in classifying GBM patients into risk groups and refine prognosis estimates to better inform treatment decisions or stratification into clinical trials.
Collapse
Affiliation(s)
- Christopher Morrison
- Department of Radiation Oncology, University of Arizona, Tucson, AZ 85719, USA; (C.M.)
| | - Eric Weterings
- Department of Radiation Oncology, University of Arizona, Tucson, AZ 85719, USA; (C.M.)
| | - Nicholas Gravbrot
- College of Medicine, University of Arizona, Tucson Campus, Tucson, AZ 85724, USA; (N.G.); (M.H.)
| | - Michael Hammer
- College of Medicine, University of Arizona, Tucson Campus, Tucson, AZ 85724, USA; (N.G.); (M.H.)
- Department of Neurology, University of Arizona Genetics Core, Tucson, AZ 85724, USA
| | - Martin Weinand
- Department of Neurosurgery, University of Arizona, Tucson, AZ 85724, USA;
| | - Abhay Sanan
- Center for Neurosciences, Tucson, AZ 85719, USA;
| | - Ritu Pandey
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center Bioinformatics Shared Resource, and College of Medicine, University of Arizona, Tucson, AZ 85724, USA;
| | - Daruka Mahadevan
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA;
| | - Baldassarre Stea
- Department of Radiation Oncology, University of Arizona, Tucson, AZ 85719, USA; (C.M.)
| |
Collapse
|
3
|
Sun L, Lin S, Bi J, Yuan Z, Li Y, Wei W, Peng Y, Hu D, Han G. Evaluation of Radiotherapy Efficacy and Prognostic Analysis for Solid and Cystic Brain Metastases. Cancer Control 2024; 31:10732748241266476. [PMID: 39030537 PMCID: PMC11268021 DOI: 10.1177/10732748241266476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 07/21/2024] Open
Abstract
OBJECTIVES Brain metastases (BMs) are commonly categorized into cystic and solid. However, the difference in the prognosis of patients with either cystic or solid BMs following radiotherapy remains poorly understood. We used a retrospective design to elucidate the disparities in survival between these two patient groups undergoing radiotherapy and to identify factors influencing the overall survival (OS) of patients with BMs. METHODS This retrospective study encompasses 212 patients diagnosed with BMs. We meticulously analyzed the clinical characteristics, radiation therapy modalities, and risk factors influencing the OS among these patients, categorized by BMs type, post-brain radiation therapy. RESULTS A statistically significant difference in mOS was observed between the two cohorts (Solid vs Cystic: 23.1 vs 14.6 months). Subgroup analysis unveiled distinctions in mOS, particularly in patients with EGFR-mutant lung adenocarcinoma (Solid vs Cystic: 23.1 vs 6.43 months). The volume of BMs and the biological effective dose (BED) emerged as significantly prognostic factors for patients with cystic BMs. For patients with solid BMs, fraction dose, BED, and the number of BMs were identified as independent prognostic factors for survival. CONCLUSION Brain radiotherapy shows superior survival benefits for lung cancer patients with solid BMs compared to those with cystic BMs, particularly in EGFR-mutant lung cancer. In particular, patients receiving BED ≥60 Gy have a more favorable prognosis than those receiving BED <60 Gy, regardless of the type of BM (solid or cystic) in lung cancer.
Collapse
Affiliation(s)
- Lu Sun
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shouyu Lin
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianping Bi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Yuan
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Ying Li
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wei
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Peng
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Desheng Hu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guang Han
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Häger W, Toma-Dașu I, Astaraki M, Lazzeroni M. Overall survival prediction for high-grade glioma patients using mathematical modeling of tumor cell infiltration. Phys Med 2023; 113:102669. [PMID: 37603907 DOI: 10.1016/j.ejmp.2023.102669] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023] Open
Abstract
PURPOSE This study aimed at applying a mathematical framework for the prediction of high-grade gliomas (HGGs) cell invasion into normal tissues for guiding the clinical target delineation, and at investigating the possibility of using tumor infiltration maps for patient overall survival (OS) prediction. MATERIAL & METHODS A model describing tumor infiltration into normal tissue was applied to 93 HGG cases. Tumor infiltration maps and corresponding isocontours with different cell densities were produced. ROC curves were used to seek correlations between the patient OS and the volume encompassed by a particular isocontour. Area-Under-the-Curve (AUC) values were used to determine the isocontour having the highest predictive ability. The optimal cut-off volume, having the highest sensitivity and specificity, for each isocontour was used to divide the patients in two groups for a Kaplan-Meier survival analysis. RESULTS The highest AUC value was obtained for the isocontour of cell densities 1000 cells/mm3 and 2000 cells/mm3, equal to 0.77 (p < 0.05). Correlation with the GTV yielded an AUC of 0.73 (p < 0.05). The Kaplan-Meier survival analysis using the 1000 cells/mm3 isocontour and the ROC optimal cut-off volume for patient group selection rendered a hazard ratio (HR) of 2.7 (p < 0.05), while the GTV rendered a HR = 1.6 (p < 0.05). CONCLUSION The simulated tumor cell invasion is a stronger predictor of overall survival than the segmented GTV, indicating the importance of using mathematical models for cell invasion to assist in the definition of the target for HGG patients.
Collapse
Affiliation(s)
- Wille Häger
- Department of Physics, Stockholm University, Stockholm, Sweden; Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden.
| | - Iuliana Toma-Dașu
- Department of Physics, Stockholm University, Stockholm, Sweden; Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Mehdi Astaraki
- Department of Biomedical Engineering and Health Systems, Royal Institute of Technology, Huddinge, Sweden; Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Marta Lazzeroni
- Department of Physics, Stockholm University, Stockholm, Sweden; Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
5
|
Choo M, Mai VH, Kim HS, Kim DH, Ku JL, Lee SK, Park CK, An YJ, Park S. Involvement of cell shape and lipid metabolism in glioblastoma resistance to temozolomide. Acta Pharmacol Sin 2023; 44:670-679. [PMID: 36100765 PMCID: PMC9958008 DOI: 10.1038/s41401-022-00984-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Temozolomide (TMZ) has been used as standard-of-care for glioblastoma multiforme (GBM), but the resistance to TMZ develops quickly and frequently. Thus, more studies are needed to elucidate the resistance mechanisms. In the current study, we investigated the relationship among the three important phenotypes, namely TMZ-resistance, cell shape and lipid metabolism, in GBM cells. We first observed the distinct difference in cell shapes between TMZ-sensitive (U87) and resistant (U87R) GBM cells. We then conducted NMR-based lipid metabolomics, which revealed a significant increase in cholesterol and fatty acid synthesis as well as lower lipid unsaturation in U87R cells. Consistent with the lipid changes, U87R cells exhibited significantly lower membrane fluidity. The transcriptomic analysis demonstrated that lipid synthesis pathways through SREBP were upregulated in U87R cells, which was confirmed at the protein level. Fatostatin, an SREBP inhibitor, and other lipid pathway inhibitors (C75, TOFA) exhibited similar or more potent inhibition on U87R cells compared to sensitive U87 cells. The lower lipid unsaturation ratio, membrane fluidity and higher fatostatin sensitivity were all recapitulated in patient-derived TMZ-resistant primary cells. The observed ternary relationship among cell shape, lipid composition, and TMZ-resistance may be applicable to other drug-resistance cases. SREBP and fatostatin are suggested as a promising target-therapeutic agent pair for drug-resistant glioblastoma.
Collapse
Affiliation(s)
- Munki Choo
- Natural Product Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Van-Hieu Mai
- Natural Product Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Han Sun Kim
- Natural Product Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Dong-Hwa Kim
- Natural Product Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Sang Kook Lee
- Natural Product Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Chul-Kee Park
- Department of Neurosurgery, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Yong Jin An
- Natural Product Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Korea.
| | - Sunghyouk Park
- Natural Product Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
6
|
Gherasim-Morogai N, Afrasanie VA, Gafton B, Marinca MV, Alexa-Stratulat T. Can Extended Chemotherapy Improve Glioblastoma Outcomes? A Retrospective Analysis of Survival in Real-World Patients. J Pers Med 2022; 12:jpm12101670. [PMID: 36294809 PMCID: PMC9604763 DOI: 10.3390/jpm12101670] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Standard treatment for glioblastoma multiforme (GBM) is surgery followed by radiotherapy plus concurrent chemotherapy with daily temozolomide (TMZ), and six subsequent TMZ 5/28-day cycles. Research has focused on identifying more effective alternatives to the current protocol, including extension of the number of adjuvant TMZ cycles. We performed a retrospective analysis of all GBM patients treated in our hospital (160 patients, 2011−2020). Median follow-up was 16.0 months. Analysis of prognostic factors was performed with a particular focus on the benefit of extending TMZ chemotherapy. Improved survival correlated with younger age, female gender, good performance status, absence of cognitive dysfunctions, no steroid use, and total tumor resection. Median progression-free survival (PFS) was 12 months and median overall survival (OS) was 20.0 months for the entire cohort. Median OS by adjuvant TMZ was 10.0 months if no adjuvant chemotherapy given (group 0), 15.0 months for patients that did not complete six TMZ cycles (group A), 24.0 months for those that did (group B), and 29.0 months for patients having received more than six cycles (group C) (p < 0.0001). At the three-year mark, 15.9% patients were alive in group A, 24.4% in group B and 38.1% in group C. Carefully selected GBM patients may derive benefit from extending the standard adjuvant chemotherapy beyond six TMZ cycles, but more data is required.
Collapse
Affiliation(s)
| | | | - Bogdan Gafton
- Medical Oncology Department, Regional Institute of Oncology, 700483 Iasi, Romania
- Oncology Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihai Vasile Marinca
- Medical Oncology Department, Regional Institute of Oncology, 700483 Iasi, Romania
- Oncology Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence:
| | - Teodora Alexa-Stratulat
- Medical Oncology Department, Regional Institute of Oncology, 700483 Iasi, Romania
- Oncology Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
7
|
McMahon DJ, Gleeson JP, O'Reilly S, Bambury RM. Management of newly diagnosed glioblastoma multiforme: current state of the art and emerging therapeutic approaches. Med Oncol 2022; 39:129. [PMID: 35716200 DOI: 10.1007/s12032-022-01708-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/14/2022] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme represent > 50% of primary gliomas and have five year survival rates of ~ 5%. Maximal safe surgical resection followed by radiotherapy with concurrent and adjuvant temozolomide remains the standard treatment since published by Stupp et al. (in N Engl J Med 352:987-996, 2005), with additional benefit for patients with MGMT-methylated tumors. We review the current treatment landscape and ongoing efforts to improve these outcomes. An extensive literature search of Pubmed and Google Scholar involving the search terms "glioblastoma," "glioblastoma multiforme," or "GBM" for papers published to July 2021 was conducted and papers evaluated for relevance. As well as current data that informs clinical practice, we review ongoing clinical research in both newly diagnosed and recurrent settings that provides hope for a breakthrough. The Stupp protocol remains standard of care in 2021. Addition of tumor treating fields improved mOS modestly, with benefit seen in MGMT-methylated and unmethylated cohorts and also improved time to cognitive decline but has not been widely adopted. The addition of lomustine to temozolomide, in MGMT-methylated patients, also showed a mOS benefit but further investigation is required. Other promising therapeutic strategies including anti-angiogenic therapy, targeted therapy, and immunotherapy have yet to show a survival advantage. Improvements in the multidisciplinary management, surgical techniques and equipment, early palliative care, carrier support, and psychological support may be responsible for improving survival over time. Despite promising preclinical rationale, immunotherapy and targeted therapy are struggling to impact survival. A number of ongoing clinical trials provide hope for a breakthrough.
Collapse
Affiliation(s)
- D J McMahon
- Cork University Hospital, Cork, Ireland, UK.
| | | | - S O'Reilly
- Cork University Hospital, Cork, Ireland, UK
| | | |
Collapse
|
8
|
Molinaro AM, Wiencke JK, Warrier G, Koestler DC, Chunduru P, Lee JY, Hansen HM, Lee S, Anguiano J, Rice T, Bracci PM, McCoy L, Salas LA, Christensen BC, Wrensch M, Kelsey KT, Taylor JW, Clarke JL. Interactions of Age and Blood Immune Factors and Noninvasive Prediction of Glioma Survival. J Natl Cancer Inst 2022; 114:446-457. [PMID: 34597382 PMCID: PMC8902347 DOI: 10.1093/jnci/djab195] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/30/2021] [Accepted: 09/23/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Tumor-based classification of human glioma portends patient prognosis, but considerable unexplained survival variability remains. Host factors (eg, age) also strongly influence survival times, partly reflecting a compromised immune system. How blood epigenetic measures of immune characteristics and age augment molecular classifications in glioma survival has not been investigated. We assess the prognostic impact of immune cell fractions and epigenetic age in archived blood across glioma molecular subtypes for the first time. METHODS We evaluated immune cell fractions and epigenetic age in archived blood from the University of California San Francisco Adult Glioma Study, which included a training set of 197 patients with IDH-wild type, 1p19q intact, TERT wild type (IDH/1p19q/TERT-WT) glioma, an evaluation set of 350 patients with other subtypes of glioma, and 454 patients without glioma. RESULTS IDH/1p19q/TERT-WT patients had lower lymphocyte fractions (CD4+ T, CD8+ T, natural killer, and B cells) and higher neutrophil fractions than people without glioma. Recursive partitioning analysis delineated 4 statistically significantly different survival groups for patients with IDH/1p19q/TERT-WT based on an interaction between chronological age and 2 blood immune factors, CD4+ T cells, and neutrophils. Median overall survival ranged from 0.76 years (95% confidence interval = 0.55-0.99) for the worst survival group (n = 28) to 9.72 years (95% confidence interval = 6.18 to not available) for the best (n = 33). The recursive partitioning analysis also statistically significantly delineated 4 risk groups in patients with other glioma subtypes. CONCLUSIONS The delineation of different survival groups in the training and evaluation sets based on an interaction between chronological age and blood immune characteristics suggests that common host immune factors among different glioma types may affect survival. The ability of DNA methylation-based markers of immune status to capture diverse, clinically relevant information may facilitate noninvasive, personalized patient evaluation in the neuro-oncology clinic.
Collapse
Affiliation(s)
- Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Gayathri Warrier
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Pranathi Chunduru
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ji Yoon Lee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Helen M Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Sean Lee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Joaquin Anguiano
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Terri Rice
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Paige M Bracci
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Lucie McCoy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Departments of Molecular and Systems Biology and Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Karl T Kelsey
- Departments of Epidemiology and Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer L Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Travers S, Litofsky NS. Daily Lifestyle Modifications to Improve Quality of Life and Survival in Glioblastoma: A Review. Brain Sci 2021; 11:brainsci11050533. [PMID: 33922443 PMCID: PMC8146925 DOI: 10.3390/brainsci11050533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 12/21/2022] Open
Abstract
Survival in glioblastoma remains poor despite advancements in standard-of-care treatment. Some patients wish to take a more active role in their cancer treatment by adopting daily lifestyle changes to improve their quality of life or overall survival. We review the available literature through PubMed and Google Scholar to identify laboratory animal studies, human studies, and ongoing clinical trials. We discuss which health habits patients adopt and which have the most promise in glioblastoma. While results of clinical trials available on these topics are limited, dietary restrictions, exercise, use of supplements and cannabis, and smoking cessation all show some benefit in the comprehensive treatment of glioblastoma. Marital status also has an impact on survival. Further clinical trials combining standard treatments with lifestyle modifications are necessary to quantify their survival advantages.
Collapse
|
10
|
Giambattista J, Omene E, Souied O, Hsu FH. Modern Treatments for Gliomas Improve Outcome. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/1573394715666191017153045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glioma is the most common type of tumor in the central nervous system (CNS). Diagnosis
is through history, physical examination, radiology, histology and molecular profiles. Magnetic
resonance imaging is a standard workup for all CNS tumors. Multidisciplinary team management
is strongly recommended. The management of low-grade gliomas is still controversial
with regards to early surgery, radiotherapy, chemotherapy, or watchful waiting watchful waiting.
Patients with suspected high-grade gliomas should undergo an assessment by neurosurgeons for
the consideration of maximum safe resection to achieve optimal tumor debulking, and to provide
adequate tissue for histologic and molecular diagnosis. Post-operative radiotherapy and/or chemotherapy
are given depending on disease grade and patient performance. Glioblastoma are mostly
considered incurable. Treatment approaches in the elderly, pediatric population and recurrent
gliomas are discussed with the latest updates in the literature. Treatment considerations include
performance status, neurocognitive functioning, and co-morbidities. Important genetic mutations,
clinical trials and guidelines are summarized in this review.
Collapse
Affiliation(s)
| | - Egiroh Omene
- Vancouver Cancer Centre, BC Cancer Agency, Columbia, Vancouver, BC, Canada
| | - Osama Souied
- Vancouver Cancer Centre, BC Cancer Agency, Columbia, Vancouver, BC, Canada
| | - Fred H.C. Hsu
- Vancouver Cancer Centre, BC Cancer Agency, Columbia, Vancouver, BC, Canada
| |
Collapse
|
11
|
Muhammed A, Gaber MS, Elbeltagy M, El Hemaly A, Taha H, Refaat A, Zaghluol MS. Risk stratification of pediatric high-grade glioma: a newly proposed prognostic score. Childs Nerv Syst 2019; 35:2355-2362. [PMID: 31218465 DOI: 10.1007/s00381-019-04257-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 06/07/2019] [Indexed: 11/26/2022]
Abstract
OBJECTIVES High-grade glioma (HGG) is a clinical challenge. Radiation Therapy Oncology Group Recursive Partitioning Analysis (RTOG-RPA) for HGG remains the standard for assessing the prognosis of adult HGG. This study assesses the validity of the RTOG-RPA to be applied to pediatric HGG. METHODS A retrospective study was conducted on 59 pediatric HGG treated in the Children's Cancer Hospital, Egypt (CCHE) between 2007 and 2016. Several factors were studied as predictors for the disease survival, including age, gender, increased intracranial hypertension, tumor characteristics and pathology, CSF seeding, performance status, post-surgical residual, and radiation dose. The statistically significant results were integrated into a Cox-regression model to develop a prognostic risk score. RESULTS Kaplan-Meier statistics identified 13 factors that impacted the overall survival. However, Cox model showed that the histological grade IV [HR 14.2, 95%CI; (3.5-57), P < 0.0001], thalamic infiltration [HR 8.7; 95%CI; (2.9-25.9), P < 0.0001], PS ≥ 60 [HR 0.317; 95%CI; (0.13-0.776); P = 0.012], and maximum tumor dimension > 3.3 cm [HR 10.2; 95%CI; (1.58-65.89); P = 0.015] were the independent variables that predicted the overall survival. A risk score was proposed based on the presence of one or more of these factors. The median OS for the low risk (score 0-1), the intermediate-low risk (score 2), the intermediate-high risk (score 3), and the high risk (score 4) were 40, 18.5, 9.5, and 2.5 months, respectively, (P < 0.0001). CONCLUSION The proposed model and risk score could stratify pediatric patients as the RTOG-RPA do for the adults.
Collapse
Affiliation(s)
- Amr Muhammed
- Department of Clinical Oncology and Nuclear Medicine, Sohag University Hospital, Sohag, Egypt.
| | - Mohamed S Gaber
- Department of Clinical Oncology and Nuclear Medicine, Sohag University Hospital, Sohag, Egypt
| | - Mohamed Elbeltagy
- Department of Neurosurgery Children's Cancer Hospital, Egypt and Faculty of Medicine Cairo University, Cairo, Egypt
| | - Ahmed El Hemaly
- Department of Pediatric Oncology, National Cancer Institute, Cairo University and Children Cancer Hospital (CCHE), Cairo, Egypt
| | - Hala Taha
- Department of Pathology, National Cancer Institute, Cairo University and Children Cancer Hospital (CCHE), Cairo, Egypt
| | - Amal Refaat
- Radio-diagnosis Department, National Cancer Institute & Children's Cancer Hospital, Cairo, Egypt
| | - Mohamed S Zaghluol
- Department of Radiation Oncology, National Cancer Institute, Cairo University and Children Cancer Hospital (CCHE), Cairo, Egypt
| |
Collapse
|
12
|
Clinical and Molecular Recursive Partitioning Analysis of High-grade Glioma Treated With IMRT. Am J Clin Oncol 2019; 42:27-35. [PMID: 29912004 DOI: 10.1097/coc.0000000000000470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Despite multimodal treatment for high-grade gliomas, prognosis remains grim. Prior Radiation Therapy Oncology Group-Recursive Partitioning Analysis (RTOG-RPA) reports indicate based on pretreatment and treatment-related factors, a subset of patients experience a significantly improved survival. Since the development of the RTOG-RPA, high-grade gliomas have seen the widespread introduction of temozolomide and tumor oncogenetics. Here we aimed to determine whether the RTOG-RPA retained prognostic significance in the context of modern treatment, as well as generate an updated RPA incorporating both clinical and genetic variables. METHODS Patients with histologically proven glioblastoma, gliosarcoma, anaplastic astrocytoma, and anaplastic oligodendroglioma treated with intensity-modulated radiation therapy (IMRT) between 2004 and 2017 were reviewed. The primary endpoint was overall survival from date of diagnosis. Primary analysis compared actual survival rates to that expected of corresponding RTOG-RPA class. Secondary analysis utilized the rpart function to recursively partition overall survival by numerous clinical and genetic pretreatment and treatment-related variables. A tertiary analysis recursively partitioned a subset of patients in which the status of all genetic markers were known. RESULTS We identified 878 patients with histologically proven high-grade glioma treated with IMRT and 291 patients in our genetic subset. Median overall survival for the entire cohort was 14.2 months (95% confidence interval, 13.1-15.3). Applying the RTOG-RPA to our cohort validated the relative prognostic ordering of the survival classes except class II. Generating our new RPA created 7 significantly different survival classes (P<0.001, χ=584) with median survival ranging from 96.4 to 2.9 months based on age, histology, O6-methylguanine-DNA methyltransferase methylation status, radiation fractions, tumor location, radiation dose, temozolomide status, and resection status. Our second RPA of our genetic subset generated 5 significantly different survival classes (P<0.001, χ=166) with survival ranging from 65.3 to 5.6 months based on age, isocitrate dehydrogenase 1 mutation status, O6-methylguanine-DNA methyltransferase methylation status, neurological functional classification, hospitalization during IMRT, temozolomide status, and Karnofsky performance status. CONCLUSIONS The RTOG-RPA retains partial prognostic significance, however, should be updated to reflect recent advancements. This series represents a large RPA analyzing both clinical and genetic factors and generated 7 distinct survival classes. Further assessment of patients with fully available genetic markers generated 5 distinct survival classes. These survival classifications need to be validated by a prospective data set and compared against the RTOG-RPA to determine whether they provide improved prognostic power.
Collapse
|
13
|
Validation and optimization of a web-based nomogram for predicting survival of patients with newly diagnosed glioblastoma. Strahlenther Onkol 2019; 196:58-69. [PMID: 31489457 DOI: 10.1007/s00066-019-01512-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/08/2019] [Indexed: 01/05/2023]
Abstract
PURPOSE To optimize and validate a current (NRG [a newly constituted National Clinical Trials Network group through National Surgical Adjuvant Breast and Bowel Project [NSABP], the Radiation Therapy Oncology Group [RTOG] and the Gynecologic Oncology Group (GOG)]) nomogram for glioblastoma patients as part of continuous validation. METHODS We identified patients newly diagnosed with glioblastoma who were treated with temozolomide-based chemoradiotherapy between 2006 and 2016 at three large-volume hospitals. The extent of resection was determined via postoperative MRI. The discrimination and calibration abilities of the prediction algorithm were assessed; if additional factors were identified as independent prognostic factors, updated models were developed using the data from two hospitals and were externally validated using the third hospital. Models were internally validated using cross-validation and bootstrapping. RESULTS A total of 837 patients met the eligibility criteria. The median overall survival (OS) was 20.0 (95% CI 18.5-21.5) months. The original nomogram was able to estimate the 6‑, 12-, and 24-month OS probabilities, but it slightly underestimated the OS values. In multivariable Cox regression analysis, MRI-defined total resection had a greater impact on OS than that shown by the original nomogram, and two additional factors-IDH1 mutation and tumor contacting subventricular zone-were newly identified as independent prognostic values. An updated nomogram incorporating these new variables outperformed the original nomogram (C-index at 6, 12, 24, and 36 months: 0.728, 0.688, 0.688, and 0.685, respectively) and was well calibrated. External validation using an independent cohort showed C‑indices of 0.787, 0.751, 0.719, and 0.702 at 6, 12, 24, and 36 months, respectively, and was well calibrated. CONCLUSION An updated and validated nomogram incorporating the contemporary parameters can estimate individual survival outcomes in patients with glioblastoma with better accuracy.
Collapse
|
14
|
Cohen-Inbar O. Geriatric brain tumor management part II: Glioblastoma multiforme. J Clin Neurosci 2019; 67:1-4. [DOI: 10.1016/j.jocn.2019.05.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/27/2019] [Indexed: 10/26/2022]
|
15
|
McConnell DD, Carr SB, Litofsky NS. Potential effects of nicotine on glioblastoma and chemoradiotherapy: a review. Expert Rev Neurother 2019; 19:545-555. [PMID: 31092064 DOI: 10.1080/14737175.2019.1617701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Introduction: Glioblastoma multiforme (GBM) has a poor prognosis despite maximal surgical resection with subsequent multi-modal radiation and chemotherapy. Use of tobacco products following diagnosis and during the period of treatment for non-neural tumors detrimentally affects treatment and prognosis. Approximately, 16-28% of patients with glioblastoma continue to smoke after diagnosis and during treatment. The literature is sparse for information-pertaining effects of smoking and nicotine on GBM treatment and prognosis. Areas covered: This review discusses cellular pathways involved in GBM progression that might be affected by nicotine, as well as how nicotine may contribute to resistance to treatment. Similarities of GBM pathways to those in non-neural tumors are investigated for potential effects by nicotine. English language papers were identified using PubMed, Medline and Scopus databases using a combination of keywords including but not limited to the following: nicotine, vaping, tobacco, e-cigarettes, smoking, vaping AND glioblastoma or brain cancer OR/AND temozolomide, carmustine, methotrexate, procarbazine, lomustine, vincristine, and neural tumor cell lines. Expert opinion: Understanding the impact of nicotine on treatment and resistance to chemotherapeutics should allow physicians to educate their patients with GBM with evidence-based recommendations about the effects of continuing to use nicotine-containing products after diagnosis and during treatment.
Collapse
Affiliation(s)
- Diane D McConnell
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| | - Steven B Carr
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| | - N Scott Litofsky
- a Division of Neurological Surgery , University of Missouri School of Medicine , Columbia , MO , USA
| |
Collapse
|
16
|
Gao WZ, Guo LM, Xu TQ, Yin YH, Jia F. Identification of a multidimensional transcriptome signature for survival prediction of postoperative glioblastoma multiforme patients. J Transl Med 2018; 16:368. [PMID: 30572911 PMCID: PMC6302404 DOI: 10.1186/s12967-018-1744-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Background Glioblastoma multiform (GBM) is a devastating brain tumor with maximum surgical resection, radiotherapy plus concomitant and adjuvant temozolomide (TMZ) as the standard treatment. Diverse clinicopathological and molecular features are major obstacles to accurate predict survival and evaluate the efficacy of chemotherapy or radiotherapy. Reliable prognostic biomarkers are urgently needed for postoperative GBM patients. Methods The protein coding genes (PCGs) and long non-coding RNA (lncRNA) gene expression profiles of 233 GBM postoperative patients were obtained from The Cancer Genome Atlas (TCGA), TANRIC and Gene Expression Omnibus (GEO) database. We randomly divided the TCGA set into a training (n = 76) and a test set (n = 77) and used GSE7696 (n = 80) as an independent validation set. Survival analysis and the random survival forest algorithm were performed to screen survival associated signature. Results Six PCGs (EIF2AK3, EPRS, GALE, GUCY2C, MTHFD2, RNF212) and five lncRNAs (CTD-2140B24.6, LINC02015, AC068888.1, CERNA1, LINC00618) were screened out by a risk score model and formed a PCG-lncRNA signature for its predictive power was strongest (AUC = 0.78 in the training dataset). The PCG-lncRNA signature could divide patients into high- risk or low-risk group with significantly different survival (median 7.47 vs. 18.27 months, log-rank test P < 0.001) in the training dataset. Similar result was observed in the test dataset (median 11.40 vs. 16.80 months, log-rank test P = 0.001) and the independent set (median 8.93 vs. 16.22 months, log-rank test P = 0.007). Multivariable Cox regression analysis verified that it was an independent prognostic factor for the postsurgical patients with GBM. Compared with IDH mutation status, O-(6)-methylguanine DNA methyltransferase promoter methylation status and age, the signature was proved to have a superior predictive power. And stratified analysis found that the signature could further separated postoperative GBM patients who received TMZ-chemoradiation into high- and low-risk groups in TCGA and GEO dataset. Conclusions The PCG-lncRNA signature was a novel prognostic marker to predict survival and TMZ-chemoradiation response in GBM patients after surgery. Electronic supplementary material The online version of this article (10.1186/s12967-018-1744-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei-Zhen Gao
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Lie-Mei Guo
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Tian-Qi Xu
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yu-Hua Yin
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Feng Jia
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
17
|
Huang D, Wang Y, Xu L, Chen L, Cheng M, Shi W, Xiong H, Zalli D, Luo S. GLI2 promotes cell proliferation and migration through transcriptional activation of ARHGEF16 in human glioma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:247. [PMID: 30305138 PMCID: PMC6180656 DOI: 10.1186/s13046-018-0917-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/25/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND The Hedgehog (Hh) signaling pathway plays critical roles in modulating embryogenesis and maintaining tissue homeostasis, with glioma-associated oncogene (GLI) transcription factors being the main mediators. Aberrant activation of this pathway is associated with various human malignancies including glioblastoma, although the mechanistic details are not well understood. METHODS We performed a microarray analysis of genes that are differentially expressed in glioblastoma U87 cells overexpressing GLI2A, the active form of GLI2, relative to the control cells. Chromatin immunoprecipitation and dual-luciferase assays were used to determine whether Rho guanine nucleotide exchange factor 16 (ARHGEF16) is a downstream target of GLI2. Then, transwell migration, EdU and soft-agar colony formation assays were employed to test effects of ARHGEF16 on glioma cancer cell migration and proliferation, and the effects of GLI2/ARHGEF16 signaling on tumor growth were examined in vivo. Finally, we performed yeast two-hybrid assay, Co-IP and GST-pull down to identify factors that mediate effects of ARHGEF16. RESULTS We found that ARHGEF16 mRNA level was upregulated in U87 cells overexpressing GLI2A relative to control cells. GLI2 binds to the ARHGEF16 promoter and activates gene transcription. Glioma cells U87 and U118 overexpressing ARHGEF16 showed enhanced migration and proliferation relative to the control cells, while knockdown of ARHGEF16 in H4 cells led to decreased cell proliferation compared to the control H4 cells. In contrast to the promoting effect of GLI2A overexpression on glioma xenograft growth, both GLI2 inhibition and ARHGEF16 knockdown retarded tumor growth. Cytoskeleton-associated protein 5 (CKAP5) was identified as an interaction protein of ARHGEF16, which is important for the stimulatory effects of ARHGEF16 on glioma cell migration and proliferation. CONCLUSIONS These results suggest that therapeutic strategies targeting the GLI2/ARHGEF16/CKAP5 signaling axis could inhibit glioma progression and recurrence.
Collapse
Affiliation(s)
- Dengliang Huang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Nanchang, 330006, Jiangxi, China
| | - Yiting Wang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Nanchang, 330006, Jiangxi, China
| | - Linlin Xu
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Nanchang, 330006, Jiangxi, China
| | - Limin Chen
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Nanchang, 330006, Jiangxi, China
| | - Minzhang Cheng
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Nanchang, 330006, Jiangxi, China
| | - Wei Shi
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Nanchang, 330006, Jiangxi, China
| | - Huanting Xiong
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Nanchang, 330006, Jiangxi, China
| | - Detina Zalli
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, Jiangxi, China. .,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
18
|
Navarria P, Pessina F, Cozzi L, Tomatis S, Reggiori G, Simonelli M, Santoro A, Clerici E, Franzese C, Carta G, Conti Nibali M, Bello L, Scorsetti M. Phase II study of hypofractionated radiation therapy in elderly patients with newly diagnosed glioblastoma with poor prognosis. TUMORI JOURNAL 2018; 105:47-54. [PMID: 30131010 DOI: 10.1177/0300891618792483] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE: To evaluate hypofractionated radiation therapy (HFRT) given at therapeutic effective doses in a phase II study. Endpoints were progression-free survival (PFS) rate, overall survival (OS), and incidence of toxicity. METHODS: Patients with newly diagnosed glioblastoma, age ⩾70 years, Karnofsky performance scale (KPS) score ⩽60, were enrolled. The total dose of HFRT was 52.5 Gy/15 fractions, corresponded to a biological effective dose to the tumor of 70.88 Gy. RESULTS: Thirty patients were treated, with a median age of 75 years. Concurrent and adjuvant temozolomide chemotherapy (TMZ-CHT) was administered in 7 (23.3%) and 11 (40.7%) patients received only adjuvant TMZ-CHT. The median, 6-month PFS, and 12-month PFS were 5.0 months, 43.3%, and 20%, respectively. The median, 6-month OS, and 12-month OS were 8 months, 90%, and 30%, respectively. At the last observation time, 26 patients (86.7%) were dead and 4 (13.3%) were alive. No increase in steroid drugs was required during radiotherapy treatment and a reduction was possible in 12 (40%). Patients with KPS=60, RPA V, MGMT methylated status, neurological status stable or improved after surgery and who underwent HFRT with concurrent and adjuvant CHT, had the better outcome. CONCLUSION: HFRT has proven to be feasible and effective, with limited morbidity, for selected elderly and frail patients with newly diagnosed glioblastoma. The primary objective of this study was not reached in the whole cohort but only in selected patients, who need more aggressive treatment.
Collapse
Affiliation(s)
- Pierina Navarria
- 1 Radiotherapy and Radiosurgery Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Federico Pessina
- 2 Neurosurgical Oncology Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Luca Cozzi
- 1 Radiotherapy and Radiosurgery Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy.,4 Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Stefano Tomatis
- 1 Radiotherapy and Radiosurgery Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Giacomo Reggiori
- 1 Radiotherapy and Radiosurgery Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Matteo Simonelli
- 3 Hematology and Oncology Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Armando Santoro
- 3 Hematology and Oncology Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy.,4 Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Elena Clerici
- 1 Radiotherapy and Radiosurgery Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Ciro Franzese
- 1 Radiotherapy and Radiosurgery Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Giulio Carta
- 1 Radiotherapy and Radiosurgery Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Marco Conti Nibali
- 2 Neurosurgical Oncology Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Lorenzo Bello
- 2 Neurosurgical Oncology Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy
| | - Marta Scorsetti
- 1 Radiotherapy and Radiosurgery Department, Humanitas Cancer Center and Research Hospital, Rozzano, Italy.,4 Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| |
Collapse
|
19
|
Woo P, Ho J, Lam S, Ma E, Chan D, Wong WK, Mak C, Lee M, Wong ST, Chan KY, Poon WS. A Comparative Analysis of the Usefulness of Survival Prediction Models for Patients with Glioblastoma in the Temozolomide Era: The Importance of Methylguanine Methyltransferase Promoter Methylation, Extent of Resection, and Subventricular Zone Location. World Neurosurg 2018; 115:e375-e385. [PMID: 29678708 DOI: 10.1016/j.wneu.2018.04.059] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Several survival prediction models for patients with glioblastoma have been proposed, but none is widely used. This study aims to identify the predictors of overall survival (OS) and to conduct an independent comparative analysis of 5 prediction models. METHODS Multi-institutional data from 159 patients with newly diagnosed glioblastoma who received adjuvant temozolomide concomitant chemoradiotherapy (CCRT) were collected. OS was assessed by Cox proportional hazards regression and adjusted for known prognostic factors. An independent CCRT patient cohort was used to externally validate the 1) RTOG (Radiation Therapy Oncology Group) recursive partitioning analysis (RPA) model, 2) Yang RPA model, and 3) Wee RPA model, Chaichana model, and the RTOG nomogram model. The predictive accuracy for each model at 12-month survival was determined by concordance indices. Calibration plots were performed to ascertain model prediction precision. RESULTS The median OS for patients who received CCRT was 19.0 months compared with 12.7 months for those who did not (P < 0.001). Independent predictors were: 1) subventricular zone II tumors (hazard ratio [HR], 1.6; 95% confidence interval [CI], 1.0-2.5); 2) methylguanine methyltransferase promoter methylation (HR, 0.36; 95% CI, 0.2-0.6); and 3) extent of resection of >85% (HR, 0.59; 95% CI, 0.4-0.9). For 12-month OS prediction, the RTOG nomogram model was superior to the RPA models with a c-index of 0.70. Calibration plots for 12-month survival showed that none of the models was precise, but the RTOG nomogram performed relatively better. CONCLUSIONS The RTOG nomogram best predicted 12-month OS. Methylguanine methyltransferase promoter methylation status, subventricular zone tumor location, and volumetric extent of resection should be considered when constructing prediction models.
Collapse
Affiliation(s)
- Peter Woo
- Department of Neurosurgery, Kwong Wah Hospital, Hong Kong, China.
| | - Jason Ho
- Department of Neurosurgery, Tuen Mun Hospital, Hong Kong, China
| | - Sandy Lam
- Department of Neurosurgery, Kwong Wah Hospital, Hong Kong, China
| | - Eric Ma
- Department of Neurosurgery, Kwong Wah Hospital, Hong Kong, China
| | - Danny Chan
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, Hong Kong, China
| | - Wai-Kei Wong
- Department of Neurosurgery, Princess Margaret Hospital, Hong Kong, China
| | - Calvin Mak
- Department of Neurosurgery, Queen Elizabeth Hospital, Hong Kong, China
| | - Michael Lee
- Department of Neurosurgery, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China
| | - Sui-To Wong
- Department of Neurosurgery, Tuen Mun Hospital, Hong Kong, China
| | - Kwong-Yau Chan
- Department of Neurosurgery, Kwong Wah Hospital, Hong Kong, China
| | - Wai-Sang Poon
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, Hong Kong, China
| |
Collapse
|
20
|
A 4-miRNA signature to predict survival in glioblastomas. PLoS One 2017; 12:e0188090. [PMID: 29136645 PMCID: PMC5685622 DOI: 10.1371/journal.pone.0188090] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Glioblastomas are among the most lethal cancers; however, recent advances in survival have increased the need for better prognostic markers. microRNAs (miRNAs) hold great prognostic potential being deregulated in glioblastomas and highly stable in stored tissue specimens. Moreover, miRNAs control multiple genes representing an additional level of gene regulation possibly more prognostically powerful than a single gene. The aim of the study was to identify a novel miRNA signature with the ability to separate patients into prognostic subgroups. Samples from 40 glioblastoma patients were included retrospectively; patients were comparable on all clinical aspects except overall survival enabling patients to be categorized as short-term or long-term survivors based on median survival. A miRNome screening was employed, and a prognostic profile was developed using leave-one-out cross-validation. We found that expression patterns of miRNAs; particularly the four miRNAs: hsa-miR-107_st, hsa-miR-548x_st, hsa-miR-3125_st and hsa-miR-331-3p_st could determine short- and long-term survival with a predicted accuracy of 78%. Heatmap dendrograms dichotomized glioblastomas into prognostic subgroups with a significant association to survival in univariate (HR 8.50; 95% CI 3.06–23.62; p<0.001) and multivariate analysis (HR 9.84; 95% CI 2.93–33.06; p<0.001). Similar tendency was seen in The Cancer Genome Atlas (TCGA) using a 2-miRNA signature of miR-107 and miR-331 (miR sum score), which were the only miRNAs available in TCGA. In TCGA, patients with O6-methylguanine-DNA-methyltransferase (MGMT) unmethylated tumors and low miR sum score had the shortest survival. Adjusting for age and MGMT status, low miR sum score was associated with a poorer prognosis (HR 0.66; 95% CI 0.45–0.97; p = 0.033). A Kyoto Encyclopedia of Genes and Genomes analysis predicted the identified miRNAs to regulate genes involved in cell cycle regulation and survival. In conclusion, the biology of miRNAs is complex, but the identified 4-miRNA expression pattern could comprise promising biomarkers in glioblastoma stratifying patients into short- and long-term survivors.
Collapse
|
21
|
Cordova JS, Gurbani SS, Holder CA, Olson JJ, Schreibmann E, Shi R, Guo Y, Shu HKG, Shim H, Hadjipanayis CG. Semi-Automated Volumetric and Morphological Assessment of Glioblastoma Resection with Fluorescence-Guided Surgery. Mol Imaging Biol 2017; 18:454-62. [PMID: 26463215 DOI: 10.1007/s11307-015-0900-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Glioblastoma (GBM) neurosurgical resection relies on contrast-enhanced MRI-based neuronavigation. However, it is well-known that infiltrating tumor extends beyond contrast enhancement. Fluorescence-guided surgery (FGS) using 5-aminolevulinic acid (5-ALA) was evaluated to improve extent of resection (EOR) of GBMs. Preoperative morphological tumor metrics were also assessed. PROCEDURES Thirty patients from a phase II trial evaluating 5-ALA FGS in newly diagnosed GBM were assessed. Tumors were segmented preoperatively to assess morphological features as well as postoperatively to evaluate EOR and residual tumor volume (RTV). RESULTS Median EOR and RTV were 94.3 % and 0.821 cm(3), respectively. Preoperative surface area to volume ratio and RTV were significantly associated with overall survival, even when controlling for the known survival confounders. CONCLUSIONS This study supports claims that 5-ALA FGS is helpful at decreasing tumor burden and prolonging survival in GBM. Moreover, morphological indices are shown to impact both resection and patient survival.
Collapse
Affiliation(s)
- J Scott Cordova
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Saumya S Gurbani
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Chad A Holder
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Ran Shi
- Department of Biostatistics, Emory University School of Public Health, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Ying Guo
- Department of Biostatistics, Emory University School of Public Health, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Hui-Kuo G Shu
- Department of Radiation Oncology, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Hyunsuk Shim
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA. .,Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA.
| | - Costas G Hadjipanayis
- Department of Neurosurgery, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA. .,Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA. .,Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, 10 Union Square, 5th Floor, Suite 5E, New York, NY, 10003, USA.
| |
Collapse
|
22
|
Yahara K, Ohguri T, Udono H, Yamamoto J, Tomura K, Onoda T, Imada H, Nishizawa S, Korogi Y. Radiotherapy using IMRT boosts after hyperbaric oxygen therapy with chemotherapy for glioblastoma. JOURNAL OF RADIATION RESEARCH 2017; 58:351-356. [PMID: 27864508 PMCID: PMC5440883 DOI: 10.1093/jrr/rrw105] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/07/2016] [Indexed: 05/27/2023]
Abstract
The purpose of this study was to evaluate the feasibility and efficacy of radiotherapy (RT) using intensity-modulated radiotherapy (IMRT) boosts after hyperbaric oxygen (HBO) therapy with chemotherapy in patients with glioblastoma. Twenty-four patients with glioblastoma were treated with the combined therapy, which was RT using IMRT boosts after HBO with chemotherapy, and were retrospectively analyzed. The RT protocol was as follows: first, 3D conformal RT [40 Gy/20 fractions (fr)] was delivered to the gross tumor volume (GTV) and the surrounding edema, including an additional 1.5-2.0 cm. The IMRT boost doses were then continuously delivered to the GTV plus 5 mm (28 Gy/8 fr) and the surrounding edema (16 Gy/8 fr). Each IMRT boost session was performed immediately after HBO to achieve radiosensitization. The planned RT dose was completed in all patients, while HBO therapy was terminated in one patient (4%) due to Grade 2 aural pain. The toxicities were mild, no non-hematological toxicity of Grade 3-5 was observed. The 2-year overall survival (OS) and progression-free survival rates in all patients were 46.5% and 35.4%, respectively. The median OS time was 22.1 months. In conclusion, the combined therapy of RT using IMRT boosts after HBO with chemotherapy was a feasible and promising treatment modality for patients with glioblastoma. The results justify further evaluation to clarify the benefits of this therapy.
Collapse
Affiliation(s)
- Katsuya Yahara
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Takayuki Ohguri
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Hiroki Udono
- Department of Neurosurgery, Tobata Kyoritsu Hospital, Kitakyushu, Japan, University of Occupational and Environmental Health, 2-5-1 Sawami Tobata-ku, Kitakyushu 804-0093, Japan
| | - Junkoh Yamamoto
- Department of Neurosurgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Kyosuke Tomura
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Toshihiro Onoda
- Department of Cancer Therapy Center, Tobata Kyoritsu Hospital, Kitakyushu, Japan, University of Occupational and Environmental Health, 2-5-1 Sawami Tobata-ku, Kitakyushu 804-0093, Japan
| | - Hajime Imada
- Department of Cancer Therapy Center, Tobata Kyoritsu Hospital, Kitakyushu, Japan, University of Occupational and Environmental Health, 2-5-1 Sawami Tobata-ku, Kitakyushu 804-0093, Japan
| | - Shigeru Nishizawa
- Department of Neurosurgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yukunori Korogi
- Department of Radiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| |
Collapse
|
23
|
Novel recursive partitioning analysis classification for newly diagnosed glioblastoma: A multi-institutional study highlighting the MGMT promoter methylation and IDH1 gene mutation status. Radiother Oncol 2017; 123:106-111. [PMID: 28302331 DOI: 10.1016/j.radonc.2017.02.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND PURPOSE To refine the recursive partitioning analysis (RPA) classification for glioblastoma incorporating the MGMT methylation and IDH1 mutation status. METHODS AND MATERIALS Three-hundred forty patients were treated with radiotherapy plus concurrent and adjuvant temozolomide in three tertiary-referral hospitals. MGMT methylation and IDH1 mutation status were available in all patients. Methylation of the MGMT (MGMTmeth) and mutation of IDH1 (IDH1mut) were observed in 42.4% and 6.2% of the patients, respectively. RESULTS The median follow-up for survivors and all patients was 33.2 and 20.5months, respectively. The median survival (MS) was 23.6months. RPA was performed on behalf of the results of the Cox proportional hazards model. MGMT methylation generated the initial partition (MGMTmeth vs. MGMTunmeth) in the RPA. Three final RPA classes were identified; class I=MGMTmeth/IDH1mut or MGMTmeth/IDH1wt/GTR/KPS≥90 (MS, 67.2months); class II=MGMTmeth/IDH1wt/GTR/KPS<90, MGMTmeth/IDH1wt/residual disease, MGMTunmeth/age<50, or MGMTunmeth/age≥50/GTR (MS, 24.0months); class III=MGMTunmeth/age≥50/residual disease (MS, 15.2months). CONCLUSIONS A novel RPA classification for glioblastoma was formulated highlighting the impact of MGMTmeth and IDH1mut in the temozolomide era. This model integrating pertinent molecular information can be used effectively for the patient stratification in future clinical trials. An external validation is ongoing.
Collapse
|
24
|
Smrdel U, Popovic M, Zwitter M, Bostjancic E, Zupan A, Kovac V, Glavac D, Bokal D, Jerebic J. Long-term survival in glioblastoma: methyl guanine methyl transferase (MGMT) promoter methylation as independent favourable prognostic factor. Radiol Oncol 2016; 50:394-401. [PMID: 27904447 PMCID: PMC5120572 DOI: 10.1515/raon-2015-0041] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/06/2015] [Indexed: 12/28/2022] Open
Abstract
Background In spite of significant improvement after multi-modality treatment, prognosis of most patients with glioblastoma remains poor. Standard clinical prognostic factors (age, gender, extent of surgery and performance status) do not clearly predict long-term survival. The aim of this case-control study was to evaluate immuno-histochemical and genetic characteristics of the tumour as additional prognostic factors in glioblastoma. Patients and methods Long-term survivor group were 40 patients with glioblastoma with survival longer than 30 months. Control group were 40 patients with shorter survival and matched to the long-term survivor group according to the clinical prognostic factors. All patients underwent multimodality treatment with surgery, postoperative conformal radiotherapy and temozolomide during and after radiotherapy. Biopsy samples were tested for the methylation of MGMT promoter (with methylation specific polymerase chain reaction), IDH1 (with immunohistochemistry), IDH2, CDKN2A and CDKN2B (with multiplex ligation-dependent probe amplification), and 1p and 19q mutations (with fluorescent in situ hybridization). Results Methylation of MGMT promoter was found in 95% and in 36% in the long-term survivor and control groups, respectively (p < 0.001). IDH1 R132H mutated patients had a non-significant lower risk of dying from glioblastoma (p = 0.437), in comparison to patients without this mutation. Other mutations were rare, with no significant difference between the two groups. Conclusions Molecular and genetic testing offers additional prognostic and predictive information for patients with glioblastoma. The most important finding of our analysis is that in the absence of MGMT promoter methylation, longterm survival is very rare. For patients without this mutation, alternative treatments should be explored.
Collapse
Affiliation(s)
- Uros Smrdel
- Department of Radiotherapy, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Mara Popovic
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Slovenia
| | | | - Emanuela Bostjancic
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Andrej Zupan
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Viljem Kovac
- Department of Radiotherapy, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Damjan Glavac
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Drago Bokal
- Department of Mathematics and Computer Science, Faculty of Natural Sciences and Mathematics, University of Maribor, Slovenia
| | - Janja Jerebic
- Department of Mathematics and Computer Science, Faculty of Natural Sciences and Mathematics, University of Maribor, Slovenia
| |
Collapse
|
25
|
Gately L, Collins A, Murphy M, Dowling A. Age alone is not a predictor for survival in glioblastoma. J Neurooncol 2016; 129:479-485. [PMID: 27406585 DOI: 10.1007/s11060-016-2194-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 07/03/2016] [Indexed: 10/21/2022]
Abstract
Over half of glioblastoma (GBM) cases are diagnosed in patients older than 65 years. Their median overall survival (OS) is 4-5 months, compared with 12-14 months in patients younger than 70 years. This retrospective audit aims to identify patterns of care and survival of patients diagnosed with GBM at a single institution in Melbourne, Australia. Consecutive histological diagnoses of adult primary GBM from January 2010 to December 2012 were retrospectively identified from medical records. Demographic, treatment and survival characteristics were recorded until death, with follow-up to January 1st 2015. Survival was estimated by Kaplan-Meier method. Planned, sub-group analyses were conducted using multivariate Cox proportional hazards model to identify differences between elderly and younger cohorts, as well as ECOG. 165 patients were identified (36 % aged ≥70 years). Those ≥70 years had a poorer performance status (ECOG 3-4: 27 vs 10 %, p = .005); poorer median OS (2.6 vs 11.5 months, p < .001); and were less likely to receive adjuvant treatment (no treatment: 40 vs 16 %, p < .001) compared with patients <70 years. Age was not a significant predictor of poorer os (HR 1.0; 0.99-1.03; p > .05), after adjusting for other clinical factors. Significant predictors of poorer os were poor performance status (p = .001), bilateral tumours (p = .04), biopsy only (p = .001), and no adjuvant treatment (p < .001). In patients diagnosed with GBM, those older than 70 years often present with poor performance status, are less likely to receive adjuvant treatment and have inferior os compared with younger patients. Treatment recommendations should be based on performance status/fitness, not age alone.
Collapse
Affiliation(s)
- Lucy Gately
- Department of Oncology, St Vincent's Hospital, Melbourne, Australia.
| | - Anna Collins
- Centre for Palliative Care, St Vincent's Hospital, Melbourne, Australia
| | - Michael Murphy
- Department of Neurosurgery, St Vincent's Hospital, Melbourne, Australia
| | - Anthony Dowling
- Department of Oncology, St Vincent's Hospital, Melbourne, Australia
| |
Collapse
|
26
|
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
|
27
|
Stieb S, Boss A, Wurnig MC, Özbay PS, Weiss T, Guckenberger M, Riesterer O, Rossi C. Non-parametric intravoxel incoherent motion analysis in patients with intracranial lesions: Test-retest reliability and correlation with arterial spin labeling. NEUROIMAGE-CLINICAL 2016; 11:780-788. [PMID: 27354956 PMCID: PMC4910187 DOI: 10.1016/j.nicl.2016.05.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 12/26/2022]
Abstract
Intravoxel incoherent motion (IVIM) analysis of diffusion imaging data provides biomarkers of true passive water diffusion and perfusion properties. A new IVIM algorithm with variable adjustment of the b-value threshold separating diffusion and perfusion effects was applied for cerebral tissue characterization in healthy volunteers, computation of test-retest reliability, correlation with arterial spin labeling, and assessment of applicability in a small cohort of patients with malignant intracranial masses. The main results of this study are threefold: (i) accounting for regional differences in the separation of the perfusion and the diffusion components improves the reliability of the model parameters; (ii) if differences in the b-value threshold are not accounted for, a significant tissue-dependent systematic bias of the IVIM parameters occurs; (iii) accounting for voxel-wise differences in the b-value threshold improves the correlation with CBF measurements in healthy volunteers and patients. The proposed algorithm provides a robust characterization of regional micro-vascularization and cellularity without a priori assumptions on tissue diffusion properties. The glioblastoma multiforme with its inherently high variability of tumor vascularization and tumor cell density may benefit from a non-invasive clinical characterization of diffusion and perfusion properties. The novel IVIM algorithm accounts for regional differences in the separation of the perfusion and the diffusion components. The algorithm improves the reliability of IVIM parameters. The algorithm improves the correlation with CBF in healthy volunteers and patients.
Collapse
Affiliation(s)
- Sonja Stieb
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Switzerland; Center for Proton Therapy, Paul Scherrer Institute, Villigen, Switzerland
| | - Andreas Boss
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich and University of Zurich, Switzerland
| | - Moritz C Wurnig
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich and University of Zurich, Switzerland
| | - Pinar S Özbay
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich and University of Zurich, Switzerland; Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, University Hospital Zurich and University of Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Switzerland
| | - Oliver Riesterer
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Switzerland
| | - Cristina Rossi
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich and University of Zurich, Switzerland.
| |
Collapse
|
28
|
Prognostic Value of Metabolic Tumor Volume on (11)C-Methionine PET in Predicting Progression-Free Survival in High-Grade Glioma. Nucl Med Mol Imaging 2015; 49:291-7. [PMID: 26550048 DOI: 10.1007/s13139-015-0362-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/01/2015] [Accepted: 08/04/2015] [Indexed: 10/23/2022] Open
Abstract
PURPOSE C-11 methionine (MET) PET is commonly used for diagnosing high-grade glioma (HGG). Recently, volumetric analysis has been widely applied to oncologic PET imaging. In this study, we investigated the prognostic value of metabolic tumor volume (MTV) on MET PET in HGG. METHODS A total of 30 patients with anaplastic astrocytoma (n = 12) and glioblastoma multiforme (n = 18) who underwent MET PET before treatment (surgery followed by chemo-radiotherapy) were retrospectively enrolled. Maximal tumor-to-normal brain ratio (TNRmax, maximum tumor activity divided by mean of normal tissue) and MTV (volume of tumor tissue that shows uptake >1.3-fold of mean uptake in normal tissue) were measured on MET PET. Adult patients were classified into two subgroups according to Radiation Therapy Oncology Group Recursive Partitioning Analysis (RTOG RPA) classification. Prognostic values of TNRmax, MTV and clinicopathologic factors were evaluated with regard to progression-free survival (PFS). RESULTS Median PFS of all patients was 7.9 months (range 1.0-53.8 months). In univariate analysis, MTV (cutoff 35 cm(3)) was a significant prognostic factor for PFS (P = 0.01), whereas TNRmax (cutoff 3.3) and RTOG RPA class were not (P = 0.80 and 0.61, respectively). Treatment of surgical resection exhibited a borderline significance (P = 0.06). In multivariate analysis, MTV was the only independent prognostic factor for PFS (P = 0.03). CONCLUSION MTV on MET PET is a significant and independent prognostic factor for PFS in HGG patients, whereas TNRmax is not. Thus, performing volumetric analysis of MET PET is recommended in HGG for better prognostication.
Collapse
|
29
|
Seidlitz A, Siepmann T, Löck S, Juratli T, Baumann M, Krause M. Impact of waiting time after surgery and overall time of postoperative radiochemotherapy on treatment outcome in glioblastoma multiforme. Radiat Oncol 2015; 10:172. [PMID: 26276734 PMCID: PMC4554319 DOI: 10.1186/s13014-015-0478-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/29/2015] [Indexed: 01/22/2023] Open
Abstract
Background A time factor of radiooncological treatment has been demonstrated for several tumours, most prominently for head and neck squamous cell carcinoma and lung cancer. In glioblastoma multiforme studies of the impact of postoperative waiting times before initiation of radio- or radiochemotherapy were inconclusive. Moreover analysis of the impact of overall treatment time of radiochemotherapy as well as overall duration of local treatment from surgery to the end of radiochemotherapy is lacking to date. Methods In this retrospective cohort study, we included 369 consecutive patients treated at our institution between 2001 and 2014. Inclusion criteria were histologically proven glioblastoma multiforme, age ≥ 18 years, ECOG performance status 0–2 before radiotherapy, radiotherapy or radiochemotherapy with 33 × 1.8 Gy to 59.4 Gy or with 30 × 2.0 Gy to 60 Gy. The impact of postoperative waiting time, radiation treatment time and overall duration of local treatment from surgery to the end of radiotherapy on overall (OS) and progression-free (PFS) survival were evaluated under consideration of known prognostic factors by univariate Log-rank tests and multivariate Cox-regression analysis. Results The majority of patients had received simultaneous and further adjuvant chemotherapy, mainly with temozolomide. Median survival time and 2-year OS were 18.0 months and 38.9 % after radiochemotherapy compared to 12.7 months and 12.6 % after radiotherapy alone. Median progression-free survival time was 7.5 months and PFS at 2 years was 14.3 % compared to 6.0 months and 3.3 %, respectively. Significant prognostic factors in multivariate analysis were age, resection status and application of simultaneous chemotherapy. No effect of the interval between surgery and adjuvant radiotherapy (median 27, range 11–112 days), radiation treatment time (median 45, range 40–71 days) and of overall time from surgery until the end of radiotherapy (median 54, range 71–154 days) on overall and progression-free survival was evident. Conclusion Our data do not indicate a relevant time factor in the treatment of glioblastoma multiforme in a large contemporary single-centre cohort. Although this study was limited by its retrospective nature, its results indicate that short delays of postoperative radiochemotherapy, e.g. for screening of a patient for a clinical trial, may be uncritical.
Collapse
Affiliation(s)
- Annekatrin Seidlitz
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany. .,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany.
| | - Timo Siepmann
- Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Germany. .,Department of Neurology and Department of Psychotherapy and Psychosomatic Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Steffen Löck
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
| | - Tareq Juratli
- Department of Neurosurgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Michael Baumann
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany. .,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Dresden, Germany.
| | - Mechthild Krause
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany. .,OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany. .,German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Dresden, Germany.
| |
Collapse
|
30
|
Abstract
The incidence of glioblastoma (GBM) has been increasing over the past several decades with majority of this increase occurring in patients older than 70 years. In spite of the growing body of evidence in this area, it is still unclear as to the optimal management of elderly patients with GBM. The elderly are a heterogeneous population with a range of comorbid conditions, and functional, cognitive, and physiological changes, and ideally treatment decisions should be made in the context of a comprehensive geriatric assessment. Patients with a poor performance status or assessed as "frail" might be considered for less aggressive therapy such as hypofractionated radiotherapy or single-agent temozolomide, whereas those with a good functional status may still benefit from maximum resection followed by combined radiation and chemotherapy. Recent randomized trials suggest molecular markers such as O(6)-methylguanine-DNA-methyltransferase promoter methylation testing could help guide these decisions, particularly when considering monotherapy with temozolomide vs radiotherapy. Ongoing studies seek to clarify the role of concurrent treatment in this population. Clinical judgment and discussion with patients and families, weighing all the options, are necessary in each case. Ultimately, patients and the neuro-oncology community should be encouraged to participate in clinical trials focused specifically on caring for the elderly patient with GBM.
Collapse
Affiliation(s)
- Michelle Ferguson
- Division of Radiation Oncology, Department of Oncology, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - George Rodrigues
- Division of Radiation Oncology, Department of Oncology, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada.
| | - Jeffrey Cao
- Division of Radiation Oncology, Department of Oncology, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Glenn Bauman
- Division of Radiation Oncology, Department of Oncology, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| |
Collapse
|
31
|
Haddad P, Shazadi S, Samiei F, Kharrazi HH, Tabatabaeefar M, Rakhsha A, Faranoosh M, Torabi-Nami M, Dadras A, Liaghi A, Nafarieh L. An overview of neuro-oncology research and practice in Iran, three years with the NOSC initiative. Int J Clin Exp Med 2015; 8:3946-3955. [PMID: 26064296 PMCID: PMC4443130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/25/2015] [Indexed: 06/04/2023]
Abstract
Research and practice of neuro-oncology compiles clinical neuroscience expertise from neurosurgery, radiation oncology, neuroradiology, medical oncology, neuropathology and related disciplines to optimize planning and therapy in central nervous system malignancies. Such an interdisciplinary context prompted health-care providers from all related disciplines to establish the Neuro-Oncology Scientific Club (NOSC) in Iran and let it flourish since 3 years ago. With the advent of advanced technologies and through continued share of experience, NOSC members have tried to provide more integrated diagnoses and therapeutic care to brain tumor patients across the country. NOSC activities revolve around some key tenets including dissemination of education and updates, facilitation of institutional collaborations; data registry and patients' awareness. By virtue of recent insights on molecular characterization of brain tumors such as codeletion of chromosomes 1p and 19q in anaplastic gliomas and O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation in glioblastoma, a range of translational research is being followed within NOSC. The most recent NOSC meeting which was held in Tehran, recapitulated main advances and dealt with the current debates on functional neurosurgery, biological markers and neuroimaging, risk prediction models in high grade gliomas and clinical issues in pediatric neuro-oncology. This article gives an overview of current hotspots in neuro-oncology research and practice which are pursued within NOSC.
Collapse
Affiliation(s)
- Peiman Haddad
- Cancer Institute, Department of Radiation Oncology, School of Medicine, Tehran University of Medical Sciences Tehran, Iran
| | - Sohrab Shazadi
- Department of Neurosurgery, School of Medicine, Shahid Beheshti University of Medical Sciences Tehran, Iran
| | - Farhad Samiei
- Cancer Institute, Department of Radiation Oncology, School of Medicine, Tehran University of Medical Sciences Tehran, Iran
| | | | - Morteza Tabatabaeefar
- Department of Radiation Oncology, School of Medicine, Shahid Beheshti University of Medical Sciences Tehran, Iran
| | - Afshin Rakhsha
- Department of Radiation Oncology, School of Medicine, Shahid Beheshti University of Medical Sciences Tehran, Iran
| | - Mohammad Faranoosh
- MAHAK Pediatric Cancer Treatment and Research Center (MPCTRC) Tehran, Iran
| | - Mohammad Torabi-Nami
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences Shiraz, Iran ; Behphar Scientific Committee Tehran, Iran
| | - Ali Dadras
- Institute of Biochemistry and Biophysics (I.B.B.), University of Tehran Tehran, Iran ; Behphar Scientific Committee Tehran, Iran
| | - Atieh Liaghi
- Institute of Biochemistry and Biophysics (I.B.B.), University of Tehran Tehran, Iran ; Behphar Scientific Committee Tehran, Iran
| | | |
Collapse
|
32
|
Hunn MK, Bauer E, Wood CE, Gasser O, Dzhelali M, Ancelet LR, Mester B, Sharples KJ, Findlay MP, Hamilton DA, Hermans IF. Dendritic cell vaccination combined with temozolomide retreatment: results of a phase I trial in patients with recurrent glioblastoma multiforme. J Neurooncol 2014; 121:319-29. [DOI: 10.1007/s11060-014-1635-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 10/18/2014] [Indexed: 12/21/2022]
|
33
|
Hofer S, Rushing E, Preusser M, Marosi C. Molecular biology of high-grade gliomas: what should the clinician know? CHINESE JOURNAL OF CANCER 2013; 33:4-7. [PMID: 24325789 PMCID: PMC3905084 DOI: 10.5732/cjc.013.10218] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The current World Health Organization classification system of primary brain tumors is solely based on morphologic criteria. However, there is accumulating evidence that tumors with similar histology have distinct molecular signatures that significantly impact treatment response and survival. Recent practice-changing clinical trials have defined a role for routine assessment of O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation in glioblastoma patients, especially in the elderly, and 1p and 19q codeletions in patients with anaplastic glial tumors. Recently discovered molecular alterations including mutations in IDH-1/2, epidermal growth factor receptor (EGFR), and BRAF also have the potential to become targets for future drug development. This article aims to summarize current knowledge on the molecular biology of high-grade gliomas relevant to daily practice.
Collapse
Affiliation(s)
- Silvia Hofer
- Department of Medical Oncology, University Hospital Zürich, Zürich, Switzerland.
| | | | | | | |
Collapse
|
34
|
Li R, Tang D, Zhang J, Wu J, Wang L, Dong J. The temozolomide derivative 2T-P400 inhibits glioma growth via administration route of intravenous injection. J Neurooncol 2013; 116:25-30. [PMID: 24065569 DOI: 10.1007/s11060-013-1255-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 09/17/2013] [Indexed: 10/26/2022]
Abstract
The aim of this study is to investigate the inhibitory effects of 2T-P400, a derivative of temozolomide (TMZ), on glioma growth. SHG-44 and U373 human glioblastoma cell lines and SHG-44 cell subcutaneous and intracranial xenograft mouse models were used as the model system for these studies. Cell growth was analyzed using MTT assay. For intracranial glioma xenograft model, mouse brains were obtained and made as paraffin section for immunohistochemical staining. Tumor volume was calculated with this formula: tumor volume = length × width2/ 2. The results showed that 2T-P400 or TMZ significantly inhibits cell growth in a concentration dependent manner with the IC50 values of 12.90 ± 1.05 or 9.73 ± 2.12 μg/ml on SHG-44 cell line and 13.12 ± 0.86 or 10.13 ± 1.02 μg/ml on U373 cell line respectively. In SHG-44 cell subcutaneous xenograft model, the tumor volume of 2T-P400 or TMZ treated group was 1,062.12 ± 204.76 or 803.59 ± 110.32 mm3 respectively, which was significantly smaller than that in physiological saline (with volume of 1,968.85 ± 348.37 mm3) treated group. In intracranial xenograft model, the tumor volume of 2T-P400 or TMZ group was 6.12 ± 1.69 or 5.58 ± 1.45 mm3 respectively, significantly smaller than that in physiological saline group of 33.08 ± 6.88 mm3. Moreover, polyethylene glycol 400 (PEG400) exhibited no significant tumor growth inhibition. Our results indicated that 2T-P400 posses the same growth inhibitory effect as TMZ on glioblastoma cell lines and the subcutaneously and intracranially transplanted gliomas in xenograft mouse models. It may be a suitable alternate of TMZ for the treatment of glioma via intravenous administration route.
Collapse
Affiliation(s)
- Rujun Li
- Department of Neurosurgery, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
35
|
Ho J, Ondos J, Ning H, Smith S, Kreisl T, Iwamoto F, Sul J, Kim L, McNeil K, Krauze A, Shankavaram U, Fine HA, Camphausen K. Chemoirradiation for glioblastoma multiforme: the national cancer institute experience. PLoS One 2013; 8:e70745. [PMID: 23940635 PMCID: PMC3733728 DOI: 10.1371/journal.pone.0070745] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 06/28/2013] [Indexed: 12/02/2022] Open
Abstract
Purpose Standard treatment for glioblastoma (GBM) is surgery followed by radiation (RT) and temozolomide (TMZ). While there is variability in survival based on several established prognostic factors, the prognostic utility of other factors such as tumor size and location are not well established. Experimental Design The charts of ninety two patients with GBM treated with RT at the National Cancer Institute (NCI) between 1998 and 2012 were retrospectively reviewed. Most patients received RT with concurrent and adjuvant TMZ. Topographic locations were classified using preoperative imaging. Gross tumor volumes were contoured using treatment planning systems utilizing both pre-operative and post-operative MR imaging. Results At a median follow-up of 18.7 months, the median overall survival (OS) and progression-free survival (PFS) for all patients was 17.9 and 7.6 months. Patients with the smallest tumors had a median OS of 52.3 months compared to 16.3 months among patients with the largest tumors, P = 0.006. The patients who received bevacizumab after recurrence had a median OS of 23.3 months, compared to 16.3 months in patients who did not receive it, P = 0.0284. The median PFS and OS in patients with periventricular tumors was 5.7 and 17.5 months, versus 8.9 and 23.3 months in patients with non-periventricular tumors, P = 0.005. Conclusions Survival in our cohort was comparable to the outcome of the defining EORTC-NCIC trial establishing the use of RT+TMZ. This study also identifies several potential prognostic factors that may be useful in stratifying patients.
Collapse
Affiliation(s)
- Jennifer Ho
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John Ondos
- Radiation Management Associates, Bethesda, Maryland, United States of America
| | - Holly Ning
- Radiation Management Associates, Bethesda, Maryland, United States of America
| | - Sharon Smith
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Teri Kreisl
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fabio Iwamoto
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joohee Sul
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lyndon Kim
- Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Kate McNeil
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andra Krauze
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Howard A. Fine
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
36
|
Chaudhry NS, Shah AH, Ferraro N, Snelling BM, Bregy A, Madhavan K, Komotar RJ. Predictors of long-term survival in patients with glioblastoma multiforme: advancements from the last quarter century. Cancer Invest 2013; 31:287-308. [PMID: 23614654 DOI: 10.3109/07357907.2013.789899] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Over the last quarter century there has been significant progress toward identifying certain characteristics and patterns in GBM patients to predict survival times and outcomes. We sought to identify clinical predictors of survival in GBM patients from the past 24 years. We examined patient survival related to tumor locations, surgical treatment, postoperative course, radiotherapy, chemotherapy, patient age, GBM recurrence, imaging characteristics, serum, and molecular markers. We present predictors that may increase, decrease, or play no significant role in determining a GBM patient's long-term survival or affect the quality of life.
Collapse
Affiliation(s)
- Nauman S Chaudhry
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
The apoptosis-resistance in t-AUCB-treated glioblastoma cells depends on activation of Hsp27. J Neurooncol 2012; 110:187-94. [DOI: 10.1007/s11060-012-0963-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 08/08/2012] [Indexed: 10/28/2022]
|
38
|
Scoccianti S, Magrini SM, Ricardi U, Detti B, Krengli M, Parisi S, Bertoni F, Sotti G, Cipressi S, Tombolini V, Dall'oglio S, Lioce M, Saieva C, Buglione M, Mantovani C, Rubino G, Muto P, Fusco V, Fariselli L, de Renzis C, Masini L, Santoni R, Pirtoli L, Biti G. Radiotherapy and temozolomide in anaplastic astrocytoma: a retrospective multicenter study by the Central Nervous System Study Group of AIRO (Italian Association of Radiation Oncology). Neuro Oncol 2012; 14:798-807. [PMID: 22539339 DOI: 10.1093/neuonc/nos081] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although the evidence for the benefit of adding temozolomide (TMZ) to radiotherapy (RT) is limited to glioblastoma patients, there is currently a trend toward treating anaplastic astrocytomas (AAs) with combined RT + TMZ. The aim of the present study was to describe the patterns of care of patients affected by AA and, particularly, to compare the outcome of patients treated exclusively with RT with those treated with RT + TMZ. Data of 295 newly diagnosed AAs treated with postoperative RT ± TMZ in the period from 2002 to 2007 were reviewed. More than 75% of patients underwent a surgical removal. All the patients had postoperative RT; 86.1% of them were treated with 3D-conformal RT (3D-CRT). Sixty-seven percent of the entire group received postoperative chemotherapy with TMZ (n = 198). One-hundred sixty-six patients received both concomitant and sequential TMZ. Prescription of postoperative TMZ increased in the most recent period (2005-2007). One- and 4-year survival rates were 70.2% and 28.6%, respectively. No statistically significant improvement in survival was observed with the addition of TMZ to RT (P = .59). Multivariate analysis showed the statistical significance of age, presence of seizures, Recursive Partitioning Analysis classes I-III, extent of surgical removal, and 3D-CRT. Changes in the care of AA over the past years are documented. Currently there is not evidence to justify the addition of TMZ to postoperative RT for patients with newly diagnosed AA outside a clinical trial. Results of prospective and randomized trials are needed.
Collapse
Affiliation(s)
- Silvia Scoccianti
- Radiotherapy Unit, Azienda Ospedaliera Universitaria Careggi, Viale Morgagni 85, 50134 Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Scott JG, Bauchet L, Fraum TJ, Nayak L, Cooper AR, Chao ST, Suh JH, Vogelbaum MA, Peereboom DM, Zouaoui S, Mathieu-Daudé H, Fabbro-Peray P, Rigau V, Taillandier L, Abrey LE, DeAngelis LM, Shih JH, Iwamoto FM. Recursive partitioning analysis of prognostic factors for glioblastoma patients aged 70 years or older. Cancer 2012; 118:5595-600. [PMID: 22517216 DOI: 10.1002/cncr.27570] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 02/06/2012] [Accepted: 02/13/2012] [Indexed: 11/07/2022]
Abstract
BACKGROUND The most-used prognostic scheme for malignant gliomas included only patients aged 18 to 70 years. The purpose of this study was to develop a prognostic model for patients ≥70 years of age with newly diagnosed glioblastoma. METHODS A total of 437 patients ≥70 years of age with newly diagnosed glioblastoma, pooled from 2 tertiary academic institutions, was identified for recursive partitioning analysis (RPA). The resulting prognostic model, based on the final pruned RPA tree, was validated using 265 glioblastoma patients ≥70 years of age from a data set independently compiled by a French consortium. RESULTS RPA produced 9 terminal nodes, which were pruned to 4 prognostic subgroups with markedly different median survivals: subgroup I = patients <75.5 years of age who underwent surgical resection (9.3 months); subgroup II = patients ≥75.5 years of age who underwent surgical resection (6.4 months); subgroup III = patients with Karnofsky performance status of 70 to 100 who underwent biopsy only (4.6 months); and subgroup IV = patients with Karnofsky performance status <70 who underwent biopsy only (2.3 months). Application of this prognostic model to the French cohort also resulted in significantly different (P < .0001) median survivals for subgroups I (8.5 months), II (7.7 months), III (4.3 months), and IV (3.1 months). CONCLUSIONS This model divides elderly glioblastoma patients into prognostic subgroups that can be easily implemented in both the patient care and the clinical trial settings. This purely clinical prognostic model serves as a backbone for the future incorporation of the increasing number of potential molecular prognostic markers.
Collapse
Affiliation(s)
- Jacob G Scott
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
KONISHI Y, MURAGAKI Y, ISEKI H, MITSUHASHI N, OKADA Y. Patterns of Intracranial Glioblastoma Recurrence After Aggressive Surgical Resection and Adjuvant Management: Retrospective Analysis of 43 Cases. Neurol Med Chir (Tokyo) 2012; 52:577-86. [DOI: 10.2176/nmc.52.577] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Yoshiyuki KONISHI
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University
| | - Yoshihiro MURAGAKI
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University
- Department of Neurosurgery, Tokyo Women's Medical University
| | - Hiroshi ISEKI
- Faculty of Advanced Techno-Surgery, Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University
- Department of Neurosurgery, Tokyo Women's Medical University
| | - Norio MITSUHASHI
- Department of Radiation Oncology, Tokyo Women's Medical University
| | - Yoshikazu OKADA
- Department of Neurosurgery, Tokyo Women's Medical University
| |
Collapse
|
41
|
|
42
|
Cao JQ, Fisher BJ, Bauman GS, Megyesi JF, Watling CJ, Macdonald DR. Hypofractionated radiotherapy with or without concurrent temozolomide in elderly patients with glioblastoma multiforme: a review of ten-year single institutional experience. J Neurooncol 2011; 107:395-405. [PMID: 22105851 DOI: 10.1007/s11060-011-0766-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 11/14/2011] [Indexed: 11/28/2022]
Affiliation(s)
- Jeffrey Q Cao
- Department of Oncology, London Regional Cancer Program, London Health Sciences Centre, University of Western Ontario, London, ON, N6A 4L6, Canada
| | | | | | | | | | | |
Collapse
|