51
|
Richard S, Tachon G, Milin S, Wager M, Karayan-Tapon L. Dual MGMT inactivation by promoter hypermethylation and loss of the long arm of chromosome 10 in glioblastoma. Cancer Med 2020; 9:6344-6353. [PMID: 32666673 PMCID: PMC7476845 DOI: 10.1002/cam4.3217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/27/2020] [Accepted: 05/17/2020] [Indexed: 12/15/2022] Open
Abstract
Background Epigenetic inactivation of O6‐methylguanine‐methyltransferase (MGMT) gene by methylation of its promoter is predictive of Temozolomid (TMZ) response in glioblastoma (GBM). MGMT is located on chromosome 10q26 and the loss of chromosome 10q is observed in 70% of GBMs. In this study, we assessed the hypothesis that the dual inactivation of MGMT, by hypermethylation of MGMT promoter and by loss the long arm of chromosome 10 (10q), may confer greater sensitivity to TMZ. Methods A total of 149 tumor samples from patients diagnosed with GBM based on the WHO 2016 classification were included in this retrospective study between November 2016 and December 2018. Methylation status of MGMT promoter was evaluated by pyrosequencing and status of chromosome 10q was assessed by array comparative genomic hybridization. Results Glioblastoma patients with chromosome 10q loss associated with hypermethylation of MGMT promoter had significantly longer overall survival (OS) (P = .0024) and progression‐free survival (PFS) (P = .031). Indeed, median OS of patients with dual inactivation of MGMT was 21.5 months compared to 12 months and 8.1 months for groups with single MGMT inactivation by hypermethylation and by 10q loss, respectively. The group with no MGMT inactivation had 9.5 months OS. Moreover, all long‐term survivors with persistent response to TMZ treatment (OS ≥ 30 months) displayed dual inactivation of MGMT. Conclusions Our data suggest that the molecular subgroup characterized by the dual inactivation of MGMT receives greater benefit from TMZ treatment. The results of our study may be of immediate clinical interest since chromosome 10q status and methylation of MGMT promoter are commonly determined in routine practice.
Collapse
Affiliation(s)
- Sophie Richard
- Faculté de Médecine, Université de Poitiers, Poitiers, France.,Laboratoire de cancérologie biologique, CHU de Poitiers, Poitiers, France
| | - Gaëlle Tachon
- Faculté de Médecine, Université de Poitiers, Poitiers, France.,Laboratoire de cancérologie biologique, CHU de Poitiers, Poitiers, France.,Laboratoire des Neurosciences Expérimentales et Cliniques, INSERM 1084, Poitiers, France
| | - Serge Milin
- Laboratoire d'anatomopathologie, CHU de Poitiers, Poitiers, France
| | - Michel Wager
- Laboratoire de cancérologie biologique, CHU de Poitiers, Poitiers, France.,Laboratoire des Neurosciences Expérimentales et Cliniques, INSERM 1084, Poitiers, France.,CHU de Poitiers, Poitiers, France
| | - Lucie Karayan-Tapon
- Faculté de Médecine, Université de Poitiers, Poitiers, France.,Laboratoire de cancérologie biologique, CHU de Poitiers, Poitiers, France.,Laboratoire des Neurosciences Expérimentales et Cliniques, INSERM 1084, Poitiers, France
| |
Collapse
|
52
|
Leelatian N, Sinnaeve J, Mistry AM, Barone SM, Brockman AA, Diggins KE, Greenplate AR, Weaver KD, Thompson RC, Chambless LB, Mobley BC, Ihrie RA, Irish JM. Unsupervised machine learning reveals risk stratifying glioblastoma tumor cells. eLife 2020; 9:56879. [PMID: 32573435 PMCID: PMC7340505 DOI: 10.7554/elife.56879] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
A goal of cancer research is to reveal cell subsets linked to continuous clinical outcomes to generate new therapeutic and biomarker hypotheses. We introduce a machine learning algorithm, Risk Assessment Population IDentification (RAPID), that is unsupervised and automated, identifies phenotypically distinct cell populations, and determines whether these populations stratify patient survival. With a pilot mass cytometry dataset of 2 million cells from 28 glioblastomas, RAPID identified tumor cells whose abundance independently and continuously stratified patient survival. Statistical validation within the workflow included repeated runs of stochastic steps and cell subsampling. Biological validation used an orthogonal platform, immunohistochemistry, and a larger cohort of 73 glioblastoma patients to confirm the findings from the pilot cohort. RAPID was also validated to find known risk stratifying cells and features using published data from blood cancer. Thus, RAPID provides an automated, unsupervised approach for finding statistically and biologically significant cells using cytometry data from patient samples.
Collapse
Affiliation(s)
- Nalin Leelatian
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Justine Sinnaeve
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States
| | - Akshitkumar M Mistry
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Sierra M Barone
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Asa A Brockman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States
| | - Kirsten E Diggins
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States
| | - Allison R Greenplate
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Kyle D Weaver
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Reid C Thompson
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Lola B Chambless
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Bret C Mobley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Rebecca A Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| |
Collapse
|
53
|
Prognostic Value of C-Reactive Protein to Albumin Ratio in Glioblastoma Multiforme Patients Treated with Concurrent Radiotherapy and Temozolomide. Int J Inflam 2020; 2020:6947382. [PMID: 32566124 PMCID: PMC7298277 DOI: 10.1155/2020/6947382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/25/2020] [Indexed: 12/31/2022] Open
Abstract
Objective We investigated the prognostic impact of C-reactive protein to albumin ratio (CRP/Alb) on the survival outcomes of newly diagnosed glioblastoma multiforme (GBM) patients treated with radiotherapy (RT) and concurrent plus adjuvant temozolomide (TMZ). Methods The pretreatment CRP and Alb records of GBM patients who underwent RT and concurrent plus adjuvant TMZ were retrospectively analyzed. The CRP/Alb was calculated by dividing serum CRP level by serum Alb level obtained prior to RT. The availability of significant cutoff value for CRP/Alb that interacts with survival was assessed with the receiver-operating characteristic (ROC) curve analysis. The primary endpoint was the association between the CRP/Alb and the overall survival (OS). Results A total of 153 patients were analyzed. At a median follow-up of 14.7 months, median and 5-year OS rates were 16.2 months (95% CI: 12.5–19.7) and 9.5%, respectively, for the entire cohort. The ROC curve analysis identified a significant cutoff value at 0.75 point (area under the curve: 74.9%; sensitivity: 70.9%; specificity: 67.7%; P < 0.001) for CRP/Alb that interacts with OS and grouped the patients into two: CRP/Alb <0.75 (n = 61) and ≥0.75 (n = 92), respectively. Survival comparisons revealed that the CRP/Alb <0.75 was associated with a significantly superior median (22.5 versus 15.7 months; P < 0.001) and 5-year (20% versus 0%) rates than the CRP/Alb ≥0.75, which retained its independent significance in multivariate analysis (P < 0.001). Conclusion Present results suggested the pretreatment CRP/Alb as a significant and independent inflammation-based index which can be utilized for further prognostic lamination of GBM patients.
Collapse
|
54
|
Li C, Ran H, Song S, Liu W, Zou W, Jiang B, Zhao H, Shao B. Overexpression of RPN2 suppresses radiosensitivity of glioma cells by activating STAT3 signal transduction. Mol Med 2020; 26:43. [PMID: 32404045 PMCID: PMC7222591 DOI: 10.1186/s10020-020-00171-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/23/2020] [Indexed: 12/22/2022] Open
Abstract
Background Radiation therapy is the primary method of treatment for glioblastoma (GBM). Therefore, the suppression of radioresistance in GBM cells is of enormous significance. Ribophorin II (RPN2), a protein component of an N-oligosaccharyl transferase complex, has been associated with chemotherapy drug resistance in multiple cancers, including GBM. However, it remains unclear whether this also plays a role in radiation therapy resistance in GBM. Methods We conducted a bioinformatic analysis of RPN2 expression using the UCSC Cancer Genomics Browser and GEPIA database and performed an immunohistochemical assessment of RPN2 expression in biopsy specimens from 34 GBM patients who had received radiation-based therapy. We also studied the expression and function of RPN2 in radiation-resistant GBM cells. Results We found that RPN2 expression was upregulated in GBM tumors and correlated with poor survival. The expression of RPN2 was also higher in GBM patients with tumor recurrence, who were classified to be resistant to radiation therapy. In the radiation-resistant GBM cells, the expression of RPN2 was also higher than in the parental cells. Depletion of RPN2 in resistant cells can sensitize these cells to radiation-induced apoptosis, and overexpression of RPN2 had the reverse effect. Myeloid cell leukemia 1 (MCL1) was found to be the downstream target of RPN2, and contributed to radiation resistance in GBM cells. Furthermore, STAT3 was found to be the regulator of MCL1, which can be activated by RPN2 dysregulation. Conclusion Our study has revealed a novel function of RPN2 in radiation-resistant GBM, and has shown that MCL1 depletion or suppression could be a promising method of therapy to overcome the resistance promoted by RPN2 dysregulation.
Collapse
Affiliation(s)
- Changyu Li
- Neurosurgery, Hainan Cancer Hospital, Haikou, China
| | - Haonan Ran
- Radiotherapy Department, Hainan Cancer Hospital, Haikou, China
| | - Shaojun Song
- Neurosurgery, Hainan Cancer Hospital, Haikou, China
| | - Weisong Liu
- Head and Neck Surgery, Hainan Cancer Hospital, Haikou, China
| | - Wenhui Zou
- Neurosurgery, Hainan Cancer Hospital, Haikou, China
| | - Bei Jiang
- Hematology Department, Hainan Cancer Hospital, Haikou, China
| | - Hongmei Zhao
- Clinical Pharmacy Department, Hainan Cancer Hospital, Haikou, China
| | - Bin Shao
- Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No.199 Dazhi Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
55
|
Straube C, Kessel KA, Antoni S, Gempt J, Meyer B, Schlegel J, Schmidt-Graf F, Combs SE. A balanced score to predict survival of elderly patients newly diagnosed with glioblastoma. Radiat Oncol 2020; 15:97. [PMID: 32375830 PMCID: PMC7201994 DOI: 10.1186/s13014-020-01549-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Over the past years, several treatment regimens have been recommended for elderly patients with glioblastoma (GBM), ranging from ultrahypofractionated radiotherapy (RT) over monochemotherapy (ChT) to combined radiochemotherapy (RChT). The current guidelines recommend active treatment in elderly patients in cases with a KPS of at least 60%. We established a score for selecting patients with a very poor prognosis from patients with a better prognosis. METHODS One hundred eighty one patients ≥65 years old, histologically diagnosed with GBM, were retrospectively evaluated. Clinical characteristics were analysed for their impact on the overall survival (OS). Factors which were significant in univariate analysis (log-rank test, p < 0.05) were included in a multi-variate model (multi-variate Cox regression analysis, MVA). The 9-month OS for the significant factors after MVA (p < 0.05) was included in a prognostic score. Score sums with a median OS of < and > 6 months were summarized as Group A and B, respectively. RESULTS Age, KPS, MGMT status, the extent of resection, aphasia after surgery and motor dysfunction after surgery were significantly associated with OS on univariate analysis (p < 0.05). On MVA age (p 0.002), MGMT promotor methylation (p 0.013) and Karnofsky performance status (p 0.005) remained significant and were included in the score. Patients were divided into two groups, group A (median OS of 2.7 months) and group B (median OS of 7.8 months). The score was of prognostic significance, independent of the adjuvant treatment regimen. CONCLUSIONS The score distinguishes patients with a poor prognosis from patients with a better prognosis. Its inclusion in future retrospective or prospective trials could help enhance the comparability of results. Before its employment on a routine basis, external validation is recommended.
Collapse
Affiliation(s)
- Christoph Straube
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.
- Deutsches Konsortium für translationale Krebsforschung (dktk), Partner Site Munich, Munich, Germany.
| | - Kerstin A Kessel
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Stefanie Antoni
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- Department of Neuropathology, Technical University of Munich, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Juergen Schlegel
- Department of Neuropathology, Technical University of Munich, Munich, Germany
| | - Friederike Schmidt-Graf
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
- Deutsches Konsortium für translationale Krebsforschung (dktk), Partner Site Munich, Munich, Germany
- Department of Radiation Sciences (DRS), Institut für Strahlenmedizin (IRM), Helmholtz Zentrum München (HMGU), Neuherberg, Germany
| |
Collapse
|
56
|
Press RH, Shafer SL, Jiang R, Buchwald ZS, Abugideiri M, Tian S, Morgan TM, Behera M, Sengupta S, Voloschin AD, Olson JJ, Hasan S, Blumenthal DT, Curran WJ, Eaton BR, Shu HKG, Zhong J. Optimal timing of chemoradiotherapy after surgical resection of glioblastoma: Stratification by validated prognostic classification. Cancer 2020; 126:3255-3264. [PMID: 32342992 DOI: 10.1002/cncr.32797] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Previous studies examining the time to initiate chemoradiation (CRT) after surgical resection of glioblastoma have been conflicting. To better define the effect that the timing of adjuvant treatment may have on outcomes, the authors examined patients within the National Cancer Database (NCDB) stratified by a validated prognostic classification system. METHODS Patients with glioblastoma in the NCDB who underwent surgery and CRT from 2004 through 2013 were analyzed. Radiation Therapy Oncology Group recursive partitioning analysis (RPA) class (III, IV, V) was extrapolated for the cohort. Time intervals were grouped weekly, with weeks 4 to 5 serving as the reference category for analyses. Kaplan-Meier analysis, log-rank testing, and multivariate (MVA) Cox proportional hazards regression were performed. RESULTS In total, 30,414 patients were included. RPA classes III, IV, and V contained 5250, 20,855, and 4309 patients, respectively. On MVA, no time point after week 5 was associated with a change in overall survival for the entire cohort or for any RPA class subgroup. The periods of weeks 0 to 1 (hazard ratio [HR], 1.18; 95% CI, 1.02-1.36), >1 to 2 (HR, 1.23; 95% CI, 1.16-1.31), and >2 to 3 (HR, 1.11; 95% CI, 1.07-1.15) demonstrated slightly worse overall survival (all P < .03). The detriment to early initiation was consistent across each RPA class subgroup. CONCLUSIONS The current data provide insight into the optimal timing of CRT in patients with glioblastoma and describe RPA class-specific outcomes. In general, short delays beyond 5 weeks did not negatively affect outcomes, whereas early initiation before 3 weeks may be detrimental.
Collapse
Affiliation(s)
- Robert H Press
- Department of Radiation Oncology, New York Proton Center, New York, New York
| | - Sarah L Shafer
- Winship Research Informatics, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Renjian Jiang
- Winship Research Informatics, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Zachary S Buchwald
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Mustafa Abugideiri
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Sibo Tian
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Tiffany M Morgan
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Madhusmita Behera
- Winship Research Informatics, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Alfredo D Voloschin
- Department of Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Jeffrey J Olson
- Department of Neurosurgery, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Shaakir Hasan
- Department of Radiation Oncology, New York Proton Center, New York, New York
| | - Deborah T Blumenthal
- Department of Neuro-Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Walter J Curran
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Bree R Eaton
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Hui-Kuo G Shu
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Jim Zhong
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
57
|
Suh CH, Kim HS, Jung SC, Choi CG, Kim SJ, Kim KW. Optimized Image-Based Surrogate Endpoints in Targeted Therapies for Glioblastoma: A Systematic Review and Meta-Analysis of Phase III Randomized Controlled Trials. Korean J Radiol 2020; 21:471-482. [PMID: 32193895 PMCID: PMC7082650 DOI: 10.3348/kjr.2019.0839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 12/19/2019] [Indexed: 01/11/2023] Open
Abstract
Objective We aimed to determine the optimized image-based surrogate endpoints (IBSEs) in targeted therapies for glioblastoma through a systematic review and meta-analysis of phase III randomized controlled trials (RCTs). Materials and Methods A systematic search of OVID-MEDLINE and EMBASE for phase III RCTs on glioblastoma was performed in December 2017. Data on overall survival (OS) and IBSEs, including progression-free survival (PFS), 6-month PFS (6moPFS), 12-month PFS (12moPFS), median PFS, and objective response rate (ORR) were extracted. Weighted linear regression analysis for the hazard ratio for OS and the hazard ratios or odds ratios for IBSEs was performed. The associations between IBSEs and OS were evaluated. Subgroup analyses according to disease stage (newly diagnosed glioblastoma versus recurrent glioblastoma), types of test treatment, and types of response assessment criteria were performed. Results Twenty-three phase III RCTs published between 2000 and 2017, including 8387 patients, met the inclusion criteria. OS showed strong correlations with PFS (standardized β coefficient [R] = 0.719), 6moPFS (R = 0.647), and 12moPFS (R = 0.638). OS showed no correlations with median PFS and ORR. In subgroup analysis according to types of therapies, PFS showed the highest correlations with OS in targeted therapies for cell cycle pathways (R = 0.913) and growth factor receptors and their downstream pathways (R = 0.962). 12moPFS showed the highest correlation with OS in antiangiogenic therapy (R = 0.821). The response assessment in neuro-oncology criteria provided higher correlation coefficients between OS and IBSEs than the Macdonald criteria. Conclusion Overall, PFS is an optimized IBSE in targeted therapies for glioblastoma; however, 12moPFS is optimal in antiangiogenic therapy.
Collapse
Affiliation(s)
- Chong Hyun Suh
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| | - Seung Chai Jung
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Choong Gon Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang Joon Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Kyung Won Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
58
|
Juric V, Murphy B. Cyclin-dependent kinase inhibitors in brain cancer: current state and future directions. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:48-62. [PMID: 35582046 PMCID: PMC9094053 DOI: 10.20517/cdr.2019.105] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/11/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
Cyclin-dependent kinases (CDKs) are important regulatory enzymes in the normal physiological processes that drive cell-cycle transitions and regulate transcription. Virtually all cancers harbour genomic alterations that lead to the constitutive activation of CDKs, resulting in the proliferation of cancer cells. CDK inhibitors (CKIs) are currently in clinical use for the treatment of breast cancer, combined with endocrine therapy. In this review, we describe the potential of CKIs for the treatment of cancer with specific focus on glioblastoma (GBM), the most common and aggressive primary brain tumour in adults. Despite intense effort to combat GBM with surgery, radiation and temozolomide chemotherapy, the median survival for patients is 15 months and the majority of patients experience disease recurrence within 6-8 months of treatment onset. Novel therapeutic approaches are urgently needed for both newly diagnosed and recurrent GBM patients. In this review, we summarise the current preclinical and clinical findings emphasising that CKIs could represent an exciting novel approach for GBM treatment.
Collapse
Affiliation(s)
- Viktorija Juric
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin D02, Ireland
| | - Brona Murphy
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin D02, Ireland
| |
Collapse
|
59
|
Cao H, Li X, Wang F, Zhang Y, Xiong Y, Yang Q. Phytochemical-Mediated Glioma Targeted Treatment: Drug Resistance and Novel Delivery Systems. Curr Med Chem 2020; 27:599-629. [PMID: 31400262 DOI: 10.2174/0929867326666190809221332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 03/15/2019] [Accepted: 07/23/2019] [Indexed: 02/08/2023]
Abstract
Glioma, especially its most malignant type, Glioblastoma (GBM), is the most common and the most aggressive malignant tumour in the central nervous system. Currently, we have no specific therapies that can significantly improve its dismal prognosis. Recent studies have reported promising in vitro experimental results of several novel glioma-targeting drugs; these studies are encouraging to both researchers and patients. However, clinical trials have revealed that novel compounds that focus on a single, clear glioma genetic alteration may not achieve a satisfactory outcome or have side effects that are unbearable. Based on this consensus, phytochemicals that exhibit multiple bioactivities have recently attracted much attention. Traditional Chinese medicine and traditional Indian medicine (Ayurveda) have shown that phytocompounds inhibit glioma angiogenesis, cancer stem cells and tumour proliferation; these results suggest a novel drug therapeutic strategy. However, single phytocompounds or their direct usage may not reverse comprehensive malignancy due to poor histological penetrability or relatively unsatisfactory in vivo efficiency. Recent research that has employed temozolomide combination treatment and Nanoparticles (NPs) with phytocompounds has revealed a powerful dual-target therapy and a high blood-brain barrier penetrability, which is accompanied by low side effects and strong specific targeting. This review is focused on major phytocompounds that have contributed to glioma-targeting treatment in recent years and their role in drug resistance inhibition, as well as novel drug delivery systems for clinical strategies. Lastly, we summarize a possible research strategy for the future.
Collapse
Affiliation(s)
- Hang Cao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Feiyifan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yueqi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
60
|
Jakola AS, Sagberg LM, Gulati S, Solheim O. Advancements in predicting outcomes in patients with glioma: a surgical perspective. Expert Rev Anticancer Ther 2020; 20:167-177. [PMID: 32114857 DOI: 10.1080/14737140.2020.1735367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Diffuse glioma is a challenging neurosurgical entity. Although surgery does not provide a cure, it may greatly influence survival, brain function, and quality of life. Surgical treatment is by nature highly personalized and outcome prediction is very complex. To engage and succeed in this balancing act it is important to make best use of the information available to the neurosurgeon.Areas covered: This narrative review provides an update on advancements in predicting outcomes in patients with glioma that are relevant to neurosurgeons.Expert opinion: The classical 'gut feeling' is notoriously unreliable and better prediction strategies for patients with glioma are warranted. There are numerous tools readily available for the neurosurgeon in predicting tumor biology and survival. Predicting extent of resection, functional outcome, and quality of life remains difficult. Although machine-learning approaches are currently not readily available in daily clinical practice, there are several ongoing efforts with the use of big data sets that are likely to create new prediction models and refine the existing models.
Collapse
Affiliation(s)
- Asgeir Store Jakola
- Department of Clinical Neuroscience, Institute of Physiology and Neuroscience, Sahlgrenska Academy, Gothenburg, Sweden.,Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Neuromedicine and Movement Science, NTNU, Trondheim, Norway
| | - Lisa Millgård Sagberg
- Department of Neurosurgery, St.Olavs Hospital, Trondheim, Norway.,Department of Public Health and Nursing, NTNU, Trondheim, Norway
| | - Sasha Gulati
- Department of Neuromedicine and Movement Science, NTNU, Trondheim, Norway.,Department of Neurosurgery, St.Olavs Hospital, Trondheim, Norway
| | - Ole Solheim
- Department of Neuromedicine and Movement Science, NTNU, Trondheim, Norway.,Department of Neurosurgery, St.Olavs Hospital, Trondheim, Norway
| |
Collapse
|
61
|
Abramczyk H, Brozek-Pluska B, Jarota A, Surmacki J, Imiela A, Kopec M. A look into the use of Raman spectroscopy for brain and breast cancer diagnostics: linear and non-linear optics in cancer research as a gateway to tumor cell identity. Expert Rev Mol Diagn 2020; 20:99-115. [PMID: 32013616 DOI: 10.1080/14737159.2020.1724092] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022]
Abstract
Introduction: Currently, intensely developing of linear and non-linear optical methods for cancer detection provides a valuable tool to improve sensitivity and specificity. One of the main reasons for insufficient progress in cancer diagnostics is related to the fact that most cancer types are not only heterogeneous in their genetic composition but also reside in varying microenvironments and interact with different cell types. Until now, no technology has been fully proven for effective detecting of invasive cancer, which infiltrating the extracellular matrix.Areas covered: This review investigates the current status of Raman spectroscopy and Raman imaging for brain and breast cancer diagnostics. Moreover, the review provides a comprehensive overview of the applicability of atomic force microscopy (AFM), linear and non-linear optics in cancer research as a gateway to tumor cell identity.Expert commentary: A combination of linear and non-linear optics, particularly Raman-driven methods, has many additional advantages to identify alterations in cancer cells that are crucial for their proliferation and that distinguish them from normal cells.
Collapse
Affiliation(s)
- Halina Abramczyk
- Laboratory of Laser Molecular Spectroscopy, Lodz University of Technology, Lodz, Poland
| | - Beata Brozek-Pluska
- Laboratory of Laser Molecular Spectroscopy, Lodz University of Technology, Lodz, Poland
| | - Arkadiusz Jarota
- Laboratory of Laser Molecular Spectroscopy, Lodz University of Technology, Lodz, Poland
| | - Jakub Surmacki
- Laboratory of Laser Molecular Spectroscopy, Lodz University of Technology, Lodz, Poland
| | - Anna Imiela
- Laboratory of Laser Molecular Spectroscopy, Lodz University of Technology, Lodz, Poland
| | - Monika Kopec
- Laboratory of Laser Molecular Spectroscopy, Lodz University of Technology, Lodz, Poland
| |
Collapse
|
62
|
Combination treatment of berberine and solid lipid curcumin particles increased cell death and inhibited PI3K/Akt/mTOR pathway of human cultured glioblastoma cells more effectively than did individual treatments. PLoS One 2019; 14:e0225660. [PMID: 31841506 PMCID: PMC6913937 DOI: 10.1371/journal.pone.0225660] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
The treatment of glioblastoma is challenging for the clinician, due to its chemotherapeutic resistance. Recent findings suggest that targeting glioblastoma using anti-cancer natural polyphenols is a promising strategy. In this context, curcumin and berberine have been shown to have potent anti-cancer and anti-inflammatory effects against several malignancies. Due to the poor solubility and limited bioavailability, these compounds have limited efficacy for treating cancer. However, use of a formulation of curcumin with higher bioavailability or combining it with berberine as a co-treatment may be proving to be more efficacious against cancer. Recently, we demonstrated that solid lipid curcumin particles (SLCPs) provided more bioavailability and anti-cancer effects in cultured glioblastoma cells than did natural curcumin. Interestingly, a combination of curcumin and berberine has proven to be more effective in inhibiting growth and proliferation of cancer in the liver, breast, lung, bone and blood. However, the effect of combining these drugs for treating glioblastoma, especially with respect to its effect on activating the PI3K/Akt/mTOR pathways has not been studied. Therefore, we decided to assess the co-treatment effects of these drugs on two different glioblastoma cell lines (U-87MG and U-251MG) and neuroblastoma cell lines (SH-SY5Y) derived from human tissue. In this study, we compared single and combination (1:5) treatment of SLCP (20 μM) and berberine (100 μM) on measures of cell viability, cell death markers, levels of c-Myc and p53, along with biomarkers of the PI3K/Akt/mTOR pathways after 24–48 h of incubation. We found that co-treatment of SLCP and berberine produced more glioblastoma cell death, more DNA fragmentation, and significantly decreased ATP levels and reduced mitochondrial membrane potential than did single treatments in both glioblastoma cells lines. In addition, we observed that co-treatment inhibited the PI3K/Akt/mTOR pathway more efficiently than their single treatments. Our study suggests that combination treatments of SLCP and berberine may be a promising strategy to reduce or prevent glioblastoma growth in comparison to individual treatments using either compound.
Collapse
|
63
|
Accelerated hyperfractionated radiochemotherapy with temozolomide is equivalent to normofractionated radiochemotherapy in a retrospective analysis of patients with glioblastoma. Radiat Oncol 2019; 14:227. [PMID: 31831026 PMCID: PMC6909505 DOI: 10.1186/s13014-019-1427-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/22/2019] [Indexed: 12/28/2022] Open
Abstract
Background Current standard of treatment for newly diagnosed patients with glioblastoma (GBM) is surgical resection with adjuvant normofractionated radiotherapy (NFRT) combined with temozolomide (TMZ) chemotherapy. Hyperfractionated accelerated radiotherapy (HFRT) which was known as an option from randomized controlled trials before the temozolomide era has not been compared to the standard therapy in a randomized setting combined with TMZ. Methods Data of 152 patients with newly diagnosed GBM treated from 10/2004 until 7/2018 at a single tertiary care institution were extracted from a clinical database and retrospectively analyzed. Thirty-eight patients treated with NFRT of 60 Gy in 30 fractions (34 with simultaneous and 2 with sequential TMZ) were compared to 114 patients treated with HFRT of 54.0 Gy in 30 fraction of 1.8 Gy twice daily (109 with simultaneous and 3 with sequential TMZ). The association between treatment protocol and other variables with overall survival (OS) was assessed using univariable and multivariable Cox regression analysis; the latter was performed using variables selected by the LASSO method. Results Median overall survival (OS) was 20.3 month for the entire cohort. For patients treated with NFRT median OS was 24.4 months compared to 18.5 months in patients treated with HFRT (p = 0.131). In univariable regression analysis the use of dexamethasone during radiotherapy had a significant negative impact on OS in both patient groups, HR 2.21 (95% CI 1.47–3.31, p = 0.0001). In multivariable analysis adjusted for O6-methylguanine-DNA methyl-transferase (MGMT) promotor methylation status, salvage treatment and secondary GBM, the use of dexamethasone was still a negative prognostic factor, HR 1.95 (95% CI 1.21–3.13, p = 0.006). Positive MGMT-methylation status and salvage treatment were highly significant positive prognostic factors. There was no strong association between treatment protocol and OS (p = 0.504). Conclusions Our retrospective analysis supports the hypothesis of equivalence between HFRT and the standard protocol of treatment for GBM. For those patients who are willing to obtain the benefit of shortening the course of radiochemotherapy, HFRT may be an alternative with comparable efficacy although it was not yet tested in a large prospective randomized study against the current standard. The positive influence of salvage therapy and negative impact of concomitant use of corticosteroids should be addressed in future prospective trials. To confirm our results, we plan to perform a pooled analysis with other tertiary clinics in order to achieve better statistical reliability.
Collapse
|
64
|
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
|
65
|
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
|
66
|
Roux A, Roca P, Edjlali M, Sato K, Zanello M, Dezamis E, Gori P, Lion S, Fleury A, Dhermain F, Meder JF, Chrétien F, Lechapt E, Varlet P, Oppenheim C, Pallud J. MRI Atlas of IDH Wild-Type Supratentorial Glioblastoma: Probabilistic Maps of Phenotype, Management, and Outcomes. Radiology 2019; 293:633-643. [PMID: 31592732 DOI: 10.1148/radiol.2019190491] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Tumor location is a main prognostic parameter in patients with glioblastoma. Probabilistic MRI-based brain atlases specifying the probability of tumor location associated with important demographic, clinical, histomolecular, and management data are lacking for isocitrate dehydrogenase (IDH) wild-type glioblastomas. Purpose To correlate glioblastoma location with clinical phenotype, surgical management, and outcomes by using a probabilistic analysis in a three-dimensional (3D) MRI-based atlas. Materials and Methods This retrospective study included all adults surgically treated for newly diagnosed IDH wild-type supratentorial glioblastoma in a tertiary adult surgical neuro-oncology center (2006-2016). Semiautomated tumor segmentation and spatial normalization procedures to build a 3D MRI-based atlas were validated. The authors performed probabilistic analyses by using voxel-based lesion symptom mapping technology. The Liebermeister test was used for binary data, and the generalized linear model was used for continuous data. Results A total of 392 patients (mean age, 61 years ± 13; 233 men) were evaluated. The authors identified the preferential location of glioblastomas according to subventricular zone, age, sex, clinical presentation, revised Radiation Therapy Oncology Group-Recursive Partitioning Analysis class, Karnofsky performance status, O6-methylguanine DNA methyltransferase promoter methylation status, surgical management, and survival. The superficial location distant from the eloquent area was more likely associated with a preserved functional status at diagnosis (348 of 392 patients [89%], P < .05), a large surgical resection (173 of 392 patients [44%], P < .05), and prolonged overall survival (163 of 334 patients [49%], P < .05). In contrast, deep location and location within eloquent brain areas were more likely associated with an impaired functional status at diagnosis (44 of 392 patients [11%], P < .05), a neurologic deficit (282 of 392 patients [72%], P < .05), treatment with biopsy only (183 of 392 patients [47%], P < .05), and shortened overall survival (171 of 334 patients [51%], P < .05). Conclusion The authors identified the preferential location of isocitrate dehydrogenase wild-type glioblastomas according to parameters of interest and provided an image-based integration of multimodal information impacting survival results. This suggests the role of glioblastoma location as a surrogate and multimodal parameter integrating several known prognostic factors. © RSNA, 2019 Online supplemental material is available for this article. See also the editorial by Huang in this issue.
Collapse
Affiliation(s)
- Alexandre Roux
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Pauline Roca
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Myriam Edjlali
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Kanako Sato
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Marc Zanello
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Edouard Dezamis
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Pietro Gori
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Stéphanie Lion
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Ariane Fleury
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Frédéric Dhermain
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Jean-François Meder
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Fabrice Chrétien
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Emmanuèle Lechapt
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Pascale Varlet
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Catherine Oppenheim
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| | - Johan Pallud
- From the Department of Neurosurgery, Sainte-Anne Hospital, Paris, France (A.R., M.Z., E.D., J.P.); Paris Descartes University, Sorbonne Paris Cité, Paris, France (A.R., P.R., M.E., M.Z., J.F.M., F.C., E.L., P.V., C.O., J.P.); UMR 1266 INSERM, IMA-BRAIN, Institute of Psychiatry and Neurosciences of Paris, Paris, France (A.R., P.R., M.E., K.S., M.Z., P.G., S.L., A.F., J.F.M., P.V., C.O., J.P.); Department of Neuroradiology, Sainte-Anne Hospital, Paris, France (M.E., J.F.M., C.O.); Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan (K.S.); LTCI, Telecom ParisTech, Paris, France (P.G.); Department of Radiotherapy, Gustave Roussy University Hospital, Villejuif, France (F.D.); and Department of Neuropathology, Sainte-Anne Hospital, Paris, France (F.C., E.L., P.V.)
| |
Collapse
|
67
|
Weller J, Tzaridis T, Mack F, Steinbach JP, Schlegel U, Hau P, Krex D, Grauer O, Goldbrunner R, Bähr O, Uhl M, Seidel C, Tabatabai G, Brehmer S, Bullinger L, Galldiks N, Schaub C, Kebir S, Stummer W, Simon M, Fimmers R, Coch C, Glas M, Herrlinger U, Schäfer N. Health-related quality of life and neurocognitive functioning with lomustine-temozolomide versus temozolomide in patients with newly diagnosed, MGMT-methylated glioblastoma (CeTeG/NOA-09): a randomised, multicentre, open-label, phase 3 trial. Lancet Oncol 2019; 20:1444-1453. [PMID: 31488360 DOI: 10.1016/s1470-2045(19)30502-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The CeTeG/NOA-09 trial showed significantly longer overall survival with combined lomustine-temozolomide therapy compared with standard temozolomide for patients with glioblastoma with methylated MGMT promoter. The trial also aimed to investigate the effect of lomustine-temozolomide therapy on health-related quality of life (HRQOL) and neurocognitive function, which we report here. METHODS In this randomised, multicentre, open-label, phase 3 trial, newly diagnosed, chemoradiotherapy-naive patients with MGMT-methylated glioblastoma, aged 18-70 years, with a Karnofsky performance score of 70% or higher, were recruited and enrolled at 17 university hospitals in Germany. Patients received standard radiotherapy (60 Gy) and were randomly assigned (1:1, stratified by centre by allocating complete blocks of six to a centre, without masking) to either six 6-week courses of oral combined lomustine (100 mg/m2 on day 1) plus temozolomide (100-200 mg/m2 on days 2-6) or standard oral temozolomide (75 mg/m2 daily during radiotherapy plus six 4-week courses of temozolomide [150-200 mg/m2] on days 1-5, every 4 weeks). The primary endpoint was overall survival. HRQOL, assessed using the European Organisation for Research and Treatment of Cancer (EORTC) quality of life questionnaire core-30 and the EORTC brain cancer module (BN20); and neurocognitive function, assessed using the Mini Mental State Examination (MMSE), plus a neurocognitive test battery (NOA-07), including Trail Making Test A and B (TMT-A and B), working memory tests, and tests for lexical (Controlled Oral Word Association [COWA]) and semantic verbal fluency, were secondary endpoints analysed in the modified intention-to-treat population (mITT; all randomly assigned patients who received at least one dose of study chemotherapy). We used linear mixed-model analyses to investigate differences between treatment groups regarding HRQOL (clinically relevant ≥10 points) and MMSE scores (clinically relevant ≥3 points). The trial is registered with ClinicalTrials.gov, NCT01149109. FINDINGS Between June 17, 2011 and April 8, 2014, 141 patients were randomly assigned and 129 patients began treatment and were included in the mITT population (63 in the temozolomide and 66 in the lomustine-temozolomide group). Median follow-up for HRQOL (the item global health) was 19·4 months (IQR 7·8-38·6), for MMSE was 15·3 months (4·1-29·6), and for COWA was 11·0 months (0-27·5). We found no significant impairment regarding any item of HRQOL in the lomustine-temozolomide group (difference between the groups for global health 0·30 [95% CI -0·23 to 0·83]; p=0·26). Differences in MMSE were in favour of the temozolomide group (difference -0·11 [95% CI -0·19 to -0·03]; p=0·0058) but were not clinically relevant (1·76/30 points over 4 years). We found no significant difference between the groups in any subtest of the neurocognitive test battery (difference for COWA 0·04 [95% CI -0·01 to 0·09]; p=0·14). INTERPRETATION The absence of systematic and clinically relevant changes in HRQOL and neurocognitive function combined with the survival benefit of lomustine-temozolomide versus temozolomide alone suggests that a long-term net clinical benefit exists for patients with newly diagnosed glioblastoma with methylation of the MGMT promoter and supports the use of lomustine-temozolomide as a treatment option for these patients. FUNDING German Federal Ministry of Education and Research.
Collapse
Affiliation(s)
- Johannes Weller
- Division of Clinical Neurooncology, Department of Neurology and Center of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Theophilos Tzaridis
- Division of Clinical Neurooncology, Department of Neurology and Center of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Frederic Mack
- Division of Clinical Neurooncology, Department of Neurology and Center of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Joachim Peter Steinbach
- Dr Senckenberg Institute of Neurooncology, University of Frankfurt, Frankfurt am Main, Germany
| | - Uwe Schlegel
- Department of Neurology, University Hospital Knappschaftskrankenhaus, Ruhr-Universität Bochum, Bochum, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander Neurooncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Dietmar Krex
- Department of Neurosurgery, University of Dresden, Dresden, Germany
| | - Oliver Grauer
- Department of Neurology, University of Münster, Münster, Germany
| | | | - Oliver Bähr
- Dr Senckenberg Institute of Neurooncology, University of Frankfurt, Frankfurt am Main, Germany
| | - Martin Uhl
- Department of Neurology and Wilhelm Sander Neurooncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Clemens Seidel
- Department of Radiation Oncology, University of Leipzig, Leipzig, Germany
| | - Ghazaleh Tabatabai
- Interdisciplinary Division of Neurooncology, University of Tübingen, Tübingen, Germany
| | - Stefanie Brehmer
- Department of Neurosurgery, University of Mannheim, Mannheim, Germany
| | - Lars Bullinger
- Department of Internal Medicine, University of Ulm, Ulm, Germany
| | - Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine, Forschungszentrum Juelich, Juelich, Germany
| | - Christina Schaub
- Division of Clinical Neurooncology, Department of Neurology and Center of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Sied Kebir
- Division of Clinical Neurooncology, Department of Neurology and Center of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Walter Stummer
- Department of Neurosurgery, University of Münster, Münster, Germany
| | - Matthias Simon
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Rolf Fimmers
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Christoph Coch
- Study Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology and West German Cancer Center, German Cancer Consortium, Partner Site Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology and Center of Integrated Oncology, University Hospital Bonn, Bonn, Germany.
| | - Niklas Schäfer
- Division of Clinical Neurooncology, Department of Neurology and Center of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
68
|
Pyrosequencing versus methylation-specific PCR for assessment of MGMT methylation in tumor and blood samples of glioblastoma patients. Sci Rep 2019; 9:11125. [PMID: 31366977 PMCID: PMC6668570 DOI: 10.1038/s41598-019-47642-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/16/2019] [Indexed: 12/20/2022] Open
Abstract
Circulating biomarkers in blood may provide an interesting alternative to risky tissue biopsies in the diagnosis and follow-up of glioblastoma patients. We have assessed MGMT methylation status in blood and tissue samples from unresected glioblastoma patients who had been included in the randomized GENOM-009 trial. Paired blood and tissue samples were assessed by methylation-specific PCR (MSP) and pyrosequencing (PYR). After establishing the minimum PYR cut-off that could yield a significant difference in overall survival, we assessed the sensitivity, specificity, positive predictive value and negative predictive value (NPV) of the analyses. Methylation could be detected in cfDNA by both MSP and PYR but with low concordance with results in tissue. Sensitivity was low for both methods (31% and 38%, respectively), while specificity was higher for MSP in blood than for PYR in plasma (96% vs 76%) and NPV was similar (56 vs 57%). Concordance of results in tissue by MSP and PYR was 84.3% (P < 0.001) and correlated with outcome. We conclude that detection of cfDNA in the blood of glioblastoma patients can be an alternative when tumor tissue is not available but methods for the detection of cfDNA in blood must improve before it can replace analysis in tumor tissue.
Collapse
|
69
|
Abstract
Delineating the gross tumor volume (GTV) is a core task within radiation treatment planning. GTVs must be precisely defined irrespective of the region involved, but even more so in a sensitive area such as the brain. As precision medicine cannot exist without precision imaging, the current article aims to discuss the various imaging modalities employed in the radiation treatment planning of brain tumors.Gliomas, meningiomas, and paragangliomas are some of the most challenging tumors and the advancement in diagnostic imaging can significantly contribute to their delineation. For gliomas, irradiation based on multiparametric magnetic resonance imaging (MRI) and amino-acid positron emission tomography (PET)/computed tomography (CT) may have a higher sensitivity and specificity, which could lead to a better sparing of organs at risk and help distinguish between tumor, edema, and radiogenic alterations. Meningiomas and paragangliomas are often associated with a good prognosis. Therefore, GTV delineation according to MRI and somatostatin receptor ligand-PET/CT plays an essential role in sparing sensitive structures and maintaining a good quality of life for these patients.The combination of multiparametric MRI and PET/CT (possibly in the form of PET/MRI) presently appears to be the optimal approach for target volume delineation. The comparative efficacy of these imaging modalities has to be further evaluated in prospective trials.
Collapse
|
70
|
Amide proton transfer imaging might predict survival and IDH mutation status in high-grade glioma. Eur Radiol 2019; 29:6643-6652. [PMID: 31175415 DOI: 10.1007/s00330-019-06203-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/14/2019] [Accepted: 03/26/2019] [Indexed: 01/27/2023]
Abstract
OBJECTIVES To assess the utility of amide proton transfer (APT) imaging as an imaging biomarker to predict prognosis and molecular marker status in high-grade glioma (HGG, WHO grade III/IV). METHODS We included 71 patients with pathologically diagnosed HGG who underwent preoperative MRI with APT imaging. Overall survival (OS) and progression-free survival (PFS) according to APT signal, clinical factors, MGMT methylation status, and IDH mutation status were analyzed. Multivariate Cox regression models with and without APT signal data were constructed. Model performance was compared using the integrated AUC (iAUC). Associations between APT signals and molecular markers were assessed using the Mann-Whitney test. RESULTS High APT signal was a significant predictor for poor OS (HR = 3.21, 95% CI = 1.62-6.34) and PFS (HR = 2.22, 95% CI = 1.33-3.72) on univariate analysis. On multivariate analysis, high APT signals were an independent predictor of poor OS and PFS when clinical factors alone (OS: HR = 2.89; PFS: HR = 2.13), or in combination with molecular markers (OS: HR = 2.85; PFS: HR = 2.00), were included as covariates. The incremental prognostic value of APT signals was significant for OS and PFS. IDH-wild type was significantly associated with high APT signals (p = 0.001) when compared to IDH-mutant; however, there was no difference based on MGMT methylation status (p = 0.208). CONCLUSION High APT signal was a significant predictor of poor prognosis in HGG. APT data showed significant incremental prognostic value over clinical prognostic factors and molecular markers and may also predict IDH mutation status. KEY POINTS • Amide proton transfer (APT) imaging is a promising prognostic marker of high-grade glioma. • APT signals were significantly higher in IDH-wild type compared to IDH-mutant high-grade glioma. • APT imaging may be valuable for preoperative screening and treatment guidance.
Collapse
|
71
|
Reulen HJ, Suero Molina E, Zeidler R, Gildehaus FJ, Böning G, Gosewisch A, Stummer W. Intracavitary radioimmunotherapy of high-grade gliomas: present status and future developments. Acta Neurochir (Wien) 2019; 161:1109-1124. [PMID: 30980242 DOI: 10.1007/s00701-019-03882-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 03/20/2019] [Indexed: 02/07/2023]
Abstract
There is a distinct need for new and second-line therapies to delay or prevent local tumor regrowth after current standard of care therapy. Intracavitary radioimmunotherapy, in combination with radiotherapy, is discussed in the present review as a therapeutic strategy of high potential. We performed a systematic literature search following the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA). The available body of literature on intracavitary radioimmunotherapy (iRIT) in glioblastoma and anaplastic astrocytomas is presented. Several past and current phase I and II clinical trials, using mostly an anti-tenascin monoclonal antibody labeled with I-131, have shown median overall survival of 19-25 months in glioblastoma, while adverse events remain low. Tenascin, followed by EGFR and variants, or smaller peptides have been used as targets, and most clinical studies were performed with I-131 or Y-90 as radionuclides while only recently Re-188, I-125, and Bi-213 were applied. The pharmacokinetics of iRIT, as well as the challenges encountered with this therapy, is comprehensively discussed. This promising approach deserves further exploration in future studies by incorporating several innovative modifications.
Collapse
Affiliation(s)
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany.
| | - Reinhard Zeidler
- Helmholtz-Zentrum Munich, German Research Center for Environmental Health, Research Group Gene Vectors, Munich, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, LMU Munich, Munich, Germany
| | | | - Guido Böning
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Astrid Gosewisch
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| |
Collapse
|
72
|
Mishra S, Verma SS, Rai V, Awasthee N, Arya JS, Maiti KK, Gupta SC. Curcuma raktakanda Induces Apoptosis and Suppresses Migration in Cancer Cells: Role of Reactive Oxygen Species. Biomolecules 2019; 9:biom9040159. [PMID: 31018580 PMCID: PMC6523773 DOI: 10.3390/biom9040159] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
Although over 100 species of Curcuma are reported, only Curcuma longa is extensively studied. Curcuma raktakanda, a poorly studied species, is most commonly distributed in the Kerala state of India. For the first time, we examined the efficacy of different fractions (acetone, hexane, and ethyl acetate) of C. raktakanda against glioma, cervical, and breast cancer cell lines. As determined by mitochondrial reductase activity assay, the viability of cancer cells was decreased in a concentration-dependent manner by the three fractions. The half maximal inhibitory concentration (IC-50) values after the treatment of C-6 glioma cells for 48 h was found to be 32.97 µg/mL (acetone extract), 40.63 µg/mL (hexane extract), and 51.65 µg/mL (ethyl acetate extract). Of the three fractions, the acetone fraction was more effective. The long-term colony formation of cancer cells was significantly suppressed by the acetone fraction. Analyses using DAPI (4',6-diamidino-2-phenylindole) staining, AO/PI (acridine orange/propidium iodide) staining, DNA laddering, and sub-G1 population revealed that the acetone extract induced apoptosis in glioma cells. The extract induced reactive oxygen species generation and suppressed the expression of cell survival proteins. The migration of cancer cells was also suppressed by the acetone extract. The gas chromatography-mass spectrometry (GC-MS) analysis indicated that tetracontane, dotriacontane, hexatriacontane, pentacosane, hexacosane, and eicosane are the major components in the acetone extract. Collectively, the extract from C. raktakanda exhibited anti-carcinogenic activities in cancer cells. We are exploring whether the phytoconstituents, individually, or collectively contribute to the anti-cancer activities of C. raktakanda.
Collapse
Affiliation(s)
- Shruti Mishra
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Sumit Singh Verma
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Vipin Rai
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Nikee Awasthee
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Jayadev S Arya
- CSIR-National Institute for Interdisciplinary Science and Technology, Chemical Science and Technology Division, Organic Chemistry Section, Trivandrum-695019, India.
| | - Kaustabh K Maiti
- CSIR-National Institute for Interdisciplinary Science and Technology, Chemical Science and Technology Division, Organic Chemistry Section, Trivandrum-695019, India.
| | - Subash C Gupta
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| |
Collapse
|
73
|
Ventero MP, Fuentes-Baile M, Quereda C, Perez-Valeciano E, Alenda C, Garcia-Morales P, Esposito D, Dorado P, Manuel Barbera V, Saceda M. Radiotherapy resistance acquisition in Glioblastoma. Role of SOCS1 and SOCS3. PLoS One 2019; 14:e0212581. [PMID: 30811476 PMCID: PMC6392282 DOI: 10.1371/journal.pone.0212581] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/05/2019] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a poor prognosis type of tumour due to its resistance to chemo and radiotherapy. SOCS1 and SOCS3 have been associated with tumour progression and response to treatments in different kinds of cancers, including GBM. In this study, cell lines of IDH-wildtype GBM from primary cultures were obtained, and the role of SOCS1 and SOCS3 in the radiotherapy response was analysed. Fifty-two brain aspirates from GBM patients were processed, and six new cell lines of IDH-wildtype GBM were established. These new cell lines were characterized according to the WHO classification of CNS tumours. SOCS1 and SOCS3 expression levels were determined, at mRNA level by Q-PCR, at protein level by immunocytochemistry, and Western blot analysis. The results showed that SOCS1 and SOCS3 are overexpressed in GBM, as compared to a non-tumoral brain RNA pool. SOCS1 and SOCS3 expression were reduced by siRNA, and it was found that SOCS3 inhibition increases radioresistance in GBM cell lines, suggesting a key role of SOCS3 in radioresistant acquisition. In addition, radioresistant clonal populations obtained by selective pressure on these cell cultures also showed a significant decrease in SOCS3 expression, while SOCS1 remained unchanged. Furthermore, the induction of SOCS3 expression, under a heterologous promoter, in a radiotherapy resistant GBM cell line increased its radiosensitivity, supporting an important implication of SOCS3 in radiotherapy resistance acquisition. Finally, the treatment with TSA in the most radioresistant established cell line produced an increase in the effect of radiotherapy, that correlated with an increase in the expression of SOCS3. These effects of TSA disappeared if the increase in the expression of SOCS3 prevented with an siRNA against SOCS3. Thus, SOCS3 signal transduction pathway (JAK/STAT) could be useful to unmask new putative targets to improve radiotherapy response in GBM.
Collapse
Affiliation(s)
- Maria Paz Ventero
- Hospital General Universitario de Elche, FISABIO, Camí de l'Almazara, Elx (Alicante), Spain
| | - Maria Fuentes-Baile
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Hospital General Universitario de Alicante, C/ Maestro Alonso, Alicante (Alicante), Spain
| | - Cristina Quereda
- Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante (Alicante), Spain
| | - Elizabeth Perez-Valeciano
- Instituto de Biología Molecular y Celular, Ed. Torregaitan, Universidad Miguel Hernández, Elche (Alicante), Spain
| | - Cristina Alenda
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Hospital General Universitario de Alicante, C/ Maestro Alonso, Alicante (Alicante), Spain
| | - Pilar Garcia-Morales
- Instituto de Biología Molecular y Celular, Ed. Torregaitan, Universidad Miguel Hernández, Elche (Alicante), Spain
| | - Danilo Esposito
- Unidad de Oncología Radioterápica, ERESA, Hospital General Universitario de Elche, Camí de l'Almazara, Elx (Alicante), Spain
| | - Pilar Dorado
- Unidad de Oncología Radioterápica, ERESA, Hospital General Universitario de Elche, Camí de l'Almazara, Elx (Alicante), Spain
| | - Victor Manuel Barbera
- Hospital General Universitario de Elche, FISABIO, Camí de l'Almazara, Elx (Alicante), Spain
- Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante (Alicante), Spain
- * E-mail: (VMB); (MS)
| | - Miguel Saceda
- Hospital General Universitario de Elche, FISABIO, Camí de l'Almazara, Elx (Alicante), Spain
- Instituto de Biología Molecular y Celular, Ed. Torregaitan, Universidad Miguel Hernández, Elche (Alicante), Spain
- * E-mail: (VMB); (MS)
| |
Collapse
|
74
|
Herrlinger U, Tzaridis T, Mack F, Steinbach JP, Schlegel U, Sabel M, Hau P, Kortmann RD, Krex D, Grauer O, Goldbrunner R, Schnell O, Bähr O, Uhl M, Seidel C, Tabatabai G, Kowalski T, Ringel F, Schmidt-Graf F, Suchorska B, Brehmer S, Weyerbrock A, Renovanz M, Bullinger L, Galldiks N, Vajkoczy P, Misch M, Vatter H, Stuplich M, Schäfer N, Kebir S, Weller J, Schaub C, Stummer W, Tonn JC, Simon M, Keil VC, Nelles M, Urbach H, Coenen M, Wick W, Weller M, Fimmers R, Schmid M, Hattingen E, Pietsch T, Coch C, Glas M. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a randomised, open-label, phase 3 trial. Lancet 2019; 393:678-688. [PMID: 30782343 DOI: 10.1016/s0140-6736(18)31791-4] [Citation(s) in RCA: 338] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/12/2018] [Accepted: 07/27/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND There is an urgent need for more effective therapies for glioblastoma. Data from a previous unrandomised phase 2 trial suggested that lomustine-temozolomide plus radiotherapy might be superior to temozolomide chemoradiotherapy in newly diagnosed glioblastoma with methylation of the MGMT promoter. In the CeTeG/NOA-09 trial, we aimed to further investigate the effect of lomustine-temozolomide therapy in the setting of a randomised phase 3 trial. METHODS In this open-label, randomised, phase 3 trial, we enrolled patients from 17 German university hospitals who were aged 18-70 years, with newly diagnosed glioblastoma with methylated MGMT promoter, and a Karnofsky Performance Score of 70% and higher. Patients were randomly assigned (1:1) with a predefined SAS-generated randomisation list to standard temozolomide chemoradiotherapy (75 mg/m2 per day concomitant to radiotherapy [59-60 Gy] followed by six courses of temozolomide 150-200 mg/m2 per day on the first 5 days of the 4-week course) or to up to six courses of lomustine (100 mg/m2 on day 1) plus temozolomide (100-200 mg/m2 per day on days 2-6 of the 6-week course) in addition to radiotherapy (59-60 Gy). Because of the different schedules, patients and physicians were not masked to treatment groups. The primary endpoint was overall survival in the modified intention-to-treat population, comprising all randomly assigned patients who started their allocated chemotherapy. The prespecified test for overall survival differences was a log-rank test stratified for centre and recursive partitioning analysis class. The trial is registered with ClinicalTrials.gov, number NCT01149109. FINDINGS Between June 17, 2011, and April 8, 2014, 141 patients were randomly assigned to the treatment groups; 129 patients (63 in the temozolomide and 66 in the lomustine-temozolomide group) constituted the modified intention-to-treat population. Median overall survival was improved from 31·4 months (95% CI 27·7-47·1) with temozolomide to 48·1 months (32·6 months-not assessable) with lomustine-temozolomide (hazard ratio [HR] 0·60, 95% CI 0·35-1·03; p=0·0492 for log-rank analysis). A significant overall survival difference between groups was also found in a secondary analysis of the intention-to-treat population (n=141, HR 0·60, 95% CI 0·35-1·03; p=0·0432 for log-rank analysis). Adverse events of grade 3 or higher were observed in 32 (51%) of 63 patients in the temozolomide group and 39 (59%) of 66 patients in the lomustine-temozolomide group. There were no treatment-related deaths. INTERPRETATION Our results suggest that lomustine-temozolomide chemotherapy might improve survival compared with temozolomide standard therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter. The findings should be interpreted with caution, owing to the small size of the trial. FUNDING German Federal Ministry of Education and Research.
Collapse
Affiliation(s)
- Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany.
| | - Theophilos Tzaridis
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Frederic Mack
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | | | - Uwe Schlegel
- Department of Neurology, University Hospital Knappschaftskrankenhaus, Ruhr-Universität Bochum, Bochum, Germany
| | - Michael Sabel
- Department of Neurosurgery, University of Düsseldorf, Düsseldorf, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander Neurooncology Unit, University Hospital Regensburg, Regensburg, Germany
| | | | - Dietmar Krex
- Department of Neurosurgery, University of Dresden, Dresden, Germany
| | - Oliver Grauer
- Department of Neurology, University of Münster, Münster, Germany
| | | | - Oliver Schnell
- Department of Neurosurgery, Ludwig Maximillian University of Munich and German Cancer Consortium, Partner Site Munich, Munich, Germany; Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Oliver Bähr
- Dr Senckenberg Institute of Neurooncology, University of Frankfurt, Frankfurt, Germany
| | - Martin Uhl
- Department of Neurology and Wilhelm Sander Neurooncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Clemens Seidel
- Department of Radiation Oncology, University of Leipzig, Leipzig, Germany
| | - Ghazaleh Tabatabai
- Interdisciplinary Division of Neurooncology, University of Tübingen, Tübingen, Germany
| | - Thomas Kowalski
- Department of Neurology, University Hospital Knappschaftskrankenhaus, Ruhr-Universität Bochum, Bochum, Germany
| | - Florian Ringel
- Department of Neurosurgery, Technical University of Munich, Munich, Germany; Department of Neurosurgery, University of Mainz, Mainz, Germany
| | | | - Bogdana Suchorska
- Department of Neurosurgery, Ludwig Maximillian University of Munich and German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Stefanie Brehmer
- Department of Neurosurgery, University of Mannheim, Mannheim, Germany
| | - Astrid Weyerbrock
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Miriam Renovanz
- Department of Neurosurgery, University of Mainz, Mainz, Germany
| | - Lars Bullinger
- Department of Internal Medicine, University of Ulm, Ulm, Germany
| | - Norbert Galldiks
- Department of Neurology, University of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine (INM-3), Juelich, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité University of Berlin, Berlin, Germany
| | - Martin Misch
- Department of Neurosurgery, Charité University of Berlin, Berlin, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Moritz Stuplich
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Niklas Schäfer
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Sied Kebir
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Johannes Weller
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Christina Schaub
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Walter Stummer
- Department of Neurosurgery, University of Münster, Münster, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, Ludwig Maximillian University of Munich and German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Matthias Simon
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Vera C Keil
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Michael Nelles
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Horst Urbach
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany; Department of Neuroradiology, University of Freiburg, Freiburg, Germany
| | - Martin Coenen
- Study Centre Bonn, University Hospital Bonn, Bonn, Germany
| | - Wolfgang Wick
- Department of Neurology, University of Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Rolf Fimmers
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Matthias Schmid
- Institute for Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Elke Hattingen
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Torsten Pietsch
- Institute of Neuropathology and DGNN Brain Tumor Reference Centre, University Hospital Bonn, Bonn, Germany
| | - Christoph Coch
- Study Centre Bonn, University Hospital Bonn, Bonn, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany; Division of Clinical Neurooncology, Department of Neurology and West German Cancer Center, German Cancer Consortium, Partner Site Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
75
|
Panesar SS, D'Souza RN, Yeh FC, Fernandez-Miranda JC. Machine Learning Versus Logistic Regression Methods for 2-Year Mortality Prognostication in a Small, Heterogeneous Glioma Database. World Neurosurg X 2019; 2:100012. [PMID: 31218287 PMCID: PMC6581022 DOI: 10.1016/j.wnsx.2019.100012] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022] Open
Abstract
Background Machine learning (ML) is the application of specialized algorithms to datasets for trend delineation, categorization, or prediction. ML techniques have been traditionally applied to large, highly dimensional databases. Gliomas are a heterogeneous group of primary brain tumors, traditionally graded using histopathologic features. Recently, the World Health Organization proposed a novel grading system for gliomas incorporating molecular characteristics. We aimed to study whether ML could achieve accurate prognostication of 2-year mortality in a small, highly dimensional database of patients with glioma. Methods We applied 3 ML techniques (artificial neural networks [ANNs], decision trees [DTs], and support vector machines [SVMs]) and classical logistic regression (LR) to a dataset consisting of 76 patients with glioma of all grades. We compared the effect of applying the algorithms to the raw database versus a database where only statistically significant features were included into the algorithmic inputs (feature selection). Results Raw input consisted of 21 variables and achieved performance of accuracy/area (C.I.) under the curve of 70.7%/0.70 (49.9-88.5) for ANN, 68%/0.72 (53.4-90.4) for SVM, 66.7%/0.64 (43.6-85.0) for LR, and 65%/0.70 (51.6-89.5) for DT. Feature selected input consisted of 14 variables and achieved performance of 73.4%/0.75 (62.9-87.9) for ANN, 73.3%/0.74 (62.1-87.4) for SVM, 69.3%/0.73 (60.0-85.8) for LR, and 65.2%/0.63 (49.1-76.9) for DT. Conclusions We demonstrate that these techniques can also be applied to small, highly dimensional datasets. Our ML techniques achieved reasonable performance compared with similar studies in the literature. Although local databases may be small versus larger cancer repositories, we demonstrate that ML techniques can still be applied to their analysis; however, traditional statistical methods are of similar benefit.
Collapse
Key Words
- ANN, Artificial neural network
- AUC, Area under the curve
- CI, Confidence interval
- DT, Decision tree
- Diagnosis
- Gliomas
- LR, Logistic regression
- Logistic regression
- ML, Machine learning
- Machine learning
- NLR, Negative likelihood ratio
- NPV, Negative predictive value
- Neuro-oncology
- PLR, Positive likelihood ratio
- PPV, Positive predictive value
- Prognostication
- SVM, Support vector machine
- WHO, World Health Organization
Collapse
Affiliation(s)
- Sandip S Panesar
- Department of Neurosurgery, Stanford University, Stanford, California, USA
| | - Rhett N D'Souza
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
76
|
Ameratunga M, Pavlakis N, Wheeler H, Grant R, Simes J, Khasraw M. Anti-angiogenic therapy for high-grade glioma. Cochrane Database Syst Rev 2018; 11:CD008218. [PMID: 30480778 PMCID: PMC6516839 DOI: 10.1002/14651858.cd008218.pub4] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND This is an updated version of the original Cochrane Review published in September 2014. The most common primary brain tumours in adults are gliomas. Gliomas span a spectrum from low to high grade and are graded pathologically on a scale of one to four according to the World Health Organization (WHO) classification. High-grade glioma (HGG) carries a poor prognosis. Grade IV glioma is known as glioblastoma and carries a median survival in treated patients of about 15 months. Glioblastomas are rich in blood vessels (i.e. highly vascular) and also rich in a protein known as vascular endothelial growth factor (VEGF) that promotes new blood vessel formation (the process of angiogenesis). Anti-angiogenic agents inhibit the process of new blood vessel formation and promote regression of existing vessels. Several anti-angiogenic agents have been investigated in clinical trials, both in newly diagnosed and recurrent HGG, showing preliminary promising results. This review was undertaken to report on the benefits and harms associated with the use of anti-angiogenic agents in the treatment of HGGs. OBJECTIVES To evaluate the efficacy and toxicity of anti-angiogenic therapy in people with high-grade glioma (HGG). The intervention can be used in two broad groups: at first diagnosis as part of 'adjuvant' therapy, or in the setting of recurrent disease. SEARCH METHODS We conducted updated searches to identify published and unpublished randomised controlled trials (RCTs), including the Cochrane Central Register of Controlled Trials (CENTRAL; 2018, Issue 9), MEDLINE and Embase to October 2018. We handsearched proceedings of relevant oncology conferences up to 2018. We also searched trial registries for ongoing studies. SELECTION CRITERIA RCTs evaluating the use of anti-angiogenic therapy to treat HGG versus the same therapy without anti-angiogenic therapy. DATA COLLECTION AND ANALYSIS Review authors screened the search results and reviewed the abstracts of potentially relevant articles before retrieving the full text of eligible articles. MAIN RESULTS After a comprehensive literature search, we identified 11 eligible RCTs (3743 participants), of which 7 were included in the original review (2987 participants). There was significant design heterogeneity in the included studies, especially in the response assessment criteria used. All eligible studies were restricted to glioblastomas and there were no eligible studies evaluating other HGGs. Ten studies were available as fully published peer-reviewed manuscripts, and one study was available in abstract form. The overall risk of bias in included studies was low. This risk was based upon low rates of selection bias, detection bias, attrition bias and reporting bias. The 11 studies included in this review did not show an improvement in overall survival with the addition of anti-angiogenic therapy (pooled hazard ratio (HR) of 0.95, 95% confidence interval (CI) 0.88 to 1.02; P = 0.16; 11 studies, 3743 participants; high-certainty evidence). However, pooled analysis from 10 studies (3595 participants) showed improvement in progression-free survival with the addition of anti-angiogenic therapy (HR 0.73, 95% CI 0.68 to 0.79; P < 0.00001; high-certainty evidence).We carried out additional analyses of overall survival and progression-free survival according to treatment setting and for anti-angiogenic therapy combined with chemotherapy compared to chemotherapy alone. Pooled analysis of overall survival in either the adjuvant or recurrent setting did not show an improvement (HR 0.93, 95% CI 0.86 to 1.02; P = 0.12; 8 studies, 2833 participants; high-certainty evidence and HR 0.99, 95% CI 0.85 to 1.16; P = 0.90; 3 studies, 910 participants; moderate-certainty evidence, respectively). Pooled analysis of overall survival for anti-angiogenic therapy combined with chemotherapy compared to chemotherapy also did not clearly show an improvement (HR 0.92, 95% CI 0.85 to 1.00; P = 0.05; 11 studies, 3506 participants; low-certainty evidence). The progression-free survival in the subgroups all showed findings that demonstrated improvements in progression-free survival with the addition of anti-angiogenic therapy. Pooled analysis of progression-free survival in both the adjuvant and recurrent setting showed an improvement (HR 0.75, 95% CI 0.69 to 0.82; P < 0.00001; 8 studies, 2833 participants; high-certainty evidence and HR 0.64, 95% CI 0.54 to 0.76; P < 0.00001; 2 studies, 762 participants; moderate-certainty evidence, respectively). Pooled analysis of progression-free survival for anti-angiogenic therapy combined with chemotherapy compared to chemotherapy alone showed an improvement (HR 0.72, 95% CI 0.66 to 0.77; P < 0.00001; 10 studies, 3464 participants). Similar to trials of anti-angiogenic therapies in other solid tumours, adverse events related to this class of therapy included hypertension and proteinuria, poor wound healing, and the potential for thromboembolic events, although generally, the rate of grade 3 and 4 adverse events was low (< 14.1%) and in keeping with the literature. The impact of anti-angiogenic therapy on quality of life varied between studies. AUTHORS' CONCLUSIONS The use of anti-angiogenic therapy does not significantly improve overall survival in newly diagnosed people with glioblastoma. Thus, there is insufficient evidence to support the use of anti-angiogenic therapy for people with newly diagnosed glioblastoma at this time. Overall there is a lack of evidence of a survival advantage for anti-angiogenic therapy over chemotherapy in recurrent glioblastoma. When considering the combination anti-angiogenic therapy with chemotherapy compared with the same chemotherapy alone, there may possibly be a small improvement in overall survival. While there is strong evidence that bevacizumab (an anti-angiogenic drug) prolongs progression-free survival in newly diagnosed and recurrent glioblastoma, the impact of this on quality of life and net clinical benefit for patients remains unclear. Not addressed here is whether subsets of people with glioblastoma may benefit from anti-angiogenic therapies, nor their utility in other HGG histologies.
Collapse
Affiliation(s)
- Malaka Ameratunga
- Alfred HospitalMedical OncologyCommercial RoadMelbourneVictoriaAustralia3004
| | - Nick Pavlakis
- Royal North Shore HospitalDepartment of Medical OncologyPacific HighwaySt LeonardsNew South WalesAustralia2065
| | - Helen Wheeler
- Royal North Shore HospitalDepartment of Medical OncologyPacific HighwaySt LeonardsNew South WalesAustralia2065
| | - Robin Grant
- Western General HospitalEdinburgh Centre for Neuro‐Oncology (ECNO)Crewe RoadEdinburghScotlandUKEH4 2XU
| | - John Simes
- The University of SydneyNHMRC Clinical Trials CentreLocked Bag 77CamperdownNSWAustralia1450
| | - Mustafa Khasraw
- NHMRC Clinical Trials Centre, The University of SydneyCamperdownAustralia
| | | |
Collapse
|
77
|
Witthayanuwat S, Pesee M, Supaadirek C, Supakalin N, Thamronganantasakul K, Krusun S. Survival Analysis of Glioblastoma Multiforme. Asian Pac J Cancer Prev 2018; 19:2613-2617. [PMID: 30256068 PMCID: PMC6249474 DOI: 10.22034/apjcp.2018.19.9.2613] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Introduction: To evaluate the survival of Glioblastoma Multiforme (GBM). Material and Methods: Patients with a pathological diagnosis of Glioblastoma Multiforme (GBM) between 1 January 1994 and 30 November 2013, were retrospectively reviewed. Inclusion criteria: 1) GBM patients with confirmed pathology, 2) GBM patients were treated by multimodality therapy. Exclusion criteria: 1) GBM patients with unconfirmed pathology, 2) GBM patients with spinal involvement, 3) GBM patients with incomplete data records. Seventy-seven patients were treated by multimodality therapy such as surgery plus post-operative radiotherapy (PORT), post-operative Temozolomide (TMZ) concurrent with radiotherapy (CCRT), post-operative CCRT with adjuvant TMZ. The overall survival was calculated by the Kaplan-Meier method and the log-rank test was used to compare the survival curves. A p-value of ≤ 0.05 was considered to be statistically significant. Results: Seventy-seven patients with a median age of 53 years (range 4-76 years) showed a median survival time (MST) of 12 months. In subgroup analyses, the PORT patients revealed a MST of 11 months and 2 year overall survival (OS) rates were 17.2%, the patients with post-operative CCRT with or without adjuvant TMZ revealed a MST of 23 months and 2 year OS rates were 38.2%. The MST of patients by Recursive Partitioning Analysis (RPA), classifications III, IV, V, VI were 26.8 months, 14.2 months, 9.9 months, and 4.0 months, (p <0.001). Conclusions: The MST of the patients who had post-operative CCRT with or without adjuvant TMZ was better than the PORT group. The RPA classification can be used to predict survival. Multimodality therapy demonstrated the most effective treatment outcome. Temozolomide might be beneficial for GBM patients in order to increase survival time.
Collapse
Affiliation(s)
- Supapan Witthayanuwat
- Division of Radiotherapy, Department of Radiology, Srinagarind Hospital, Faculty of Medicine, Khon Kaen University, Thailand.
| | | | | | | | | | | |
Collapse
|
78
|
Scicchitano BM, Sorrentino S, Proietti G, Lama G, Dobrowolny G, Catizone A, Binda E, Larocca LM, Sica G. Levetiracetam enhances the temozolomide effect on glioblastoma stem cell proliferation and apoptosis. Cancer Cell Int 2018; 18:136. [PMID: 30214378 PMCID: PMC6131782 DOI: 10.1186/s12935-018-0626-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/29/2018] [Indexed: 11/10/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is a highly aggressive brain tumor in which cancer cells with stem cell-like features, called cancer stem cells (CSCs), were identified. Two CSC populations have been previously identified in GBM, one derived from the GBM area called enhanced lesion (GCSCs) and the other one from the brain area adjacent to the tumor margin (PCSCs) that greatly differ in their growth properties and tumor-initiating ability. To date the most effective chemotherapy to treat GBM is represented by alkylating agents such as temozolomide (TMZ), whose activity can be regulated by histone deacetylases (HDACs) inhibitors through the modulation of O6-methylguanine-DNA methyltransferase (MGMT) expression. Levetiracetam (LEV), a relatively new antiepileptic drug, modulates HDAC levels ultimately silencing MGMT, thus increasing TMZ effectiveness. However, an improvement in the therapeutic efficacy of TMZ is needed. Methods Cell proliferation was investigated by BrdU cell proliferation assay and by Western Blot analysis of PCNA expression. Apoptosis was evaluated by Western Blot and Immunofluorescence analysis of the cleaved Caspase-3 expression. MGMT and HDAC4 expression was analyzed by Western Blotting and Immunofluorescence. Statistical analysis was performed using the Student's t test and Mann-Whitney test. Results Here we evaluated the effect of TMZ on the proliferation rate of the IDH-wildtype GCSCs and PCSCs derived from six patients, in comparison with the effects of other drugs such as etoposide, irinotecan and carboplatin. Our results demonstrated that TMZ was less effective compared to the other agents; hence, we verified the possibility to increase the effect of TMZ by combining it with LEV. Here we show that LEV enhances the effect of TMZ on GCSCs proliferation (being less effective on PCSCs) by decreasing MGMT expression, promoting HDAC4 nuclear translocation and activating apoptotic pathway. Conclusions Although further studies are needed to determine the exact mechanism by which LEV makes GBM stem cells more sensitive to TMZ, these results suggest that the clinical therapeutic efficacy of TMZ in GBM might be enhanced by the combined treatment with LEV.
Collapse
Affiliation(s)
- Bianca Maria Scicchitano
- 1Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Silvia Sorrentino
- 1Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Gabriella Proietti
- 1Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Gina Lama
- 1Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Gabriella Dobrowolny
- 2DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Scarpa 16, 00161 Rome, Italy
| | - Angela Catizone
- 2DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Scarpa 16, 00161 Rome, Italy
| | - Elena Binda
- 3ISBReMIT-Cancer Stem Cells Unit, IRCSS Casa Sollievo della Sofferenza, Viale Padre Pio 7, 71013 San Giovanni Rotondo, FG Italy
| | - Luigi Maria Larocca
- 4Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Gigliola Sica
- 1Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| |
Collapse
|
79
|
A retrospective comparison of intensity-modulated arc therapy and 3-dimensional conformal approaches in the planning of grade 3 gliomas. JOURNAL OF RADIOTHERAPY IN PRACTICE 2018. [DOI: 10.1017/s1460396918000079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractPurposeTo evaluate the extent to which intensity-modulated arc therapy (IMAT) for high-grade gliomas is comparable with three-dimensional conformal radiotherapy (3DCRT) in relation to the dose delivered to normal brain tissue (NBT), planning target volume (PTV) conformity and the dose delivered to brainstem and optic chiasma.MethodA total of 16 randomly selected 3DCRT treatment plans of grade 3 gliomas were re-planned using an IMAT planning technique and dose–volume histograms were compared. Primary outcomes were maximum, mean, 1/3 and 2/3 doses to NBT outside the PTV. Also the maximum, mean, D50 and D20 doses to PTV. Secondary outcomes were maximum and mean doses to the brainstem and optic chiasm. Wilcoxon signed rank test was used to compare data.ResultsIMAT led to a statistically significant increase in mean dose to NBT (34·4 versus 33·3 Gy, (p=0·047) but a statistically significant reduction in maximum dose to NBT (62·7 versus 63·8 Gy, p=0·004) compared with 3DCRT. IMAT led to statistically significant reductions in maximum, D50 and D20 doses to the PTV (63·3 versus 64·7 Gy, p=0·001; 60·0 versus 60·7 Gy, p=0·001 and 60·5 versus 61·8 Gy, p=0·002, respectively). No statistically significant differences were seen in doses to brainstem and optic chiasm.ConclusionIMAT is at least comparable with 3DCRT in relation to minimising dose to NBT and ensuring good PTV conformity. Doses delivered to organs at risk using IMAT were also comparable with 3DCRT. This study supports the continued use of IMAT for the treatment of high-grade gliomas.
Collapse
|
80
|
Zhang C, Yang X, Fu C, Liu X. Combination with TMZ and miR-505 inhibits the development of glioblastoma by regulating the WNT7B/Wnt/β-catenin signaling pathway. Gene 2018; 672:172-179. [DOI: 10.1016/j.gene.2018.06.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 05/06/2018] [Accepted: 06/11/2018] [Indexed: 01/05/2023]
|
81
|
Potharaju M, Mangaleswaran B, Mathavan A, John R, Thamburaj V, Ghosh S, Ganesh S, Kalvakonda C, Loganathan M, Bapu S, Devi R, Verma RS. Body Mass Index as a Prognostic Marker in Glioblastoma Multiforme: A Clinical Outcome. Int J Radiat Oncol Biol Phys 2018; 102:204-209. [DOI: 10.1016/j.ijrobp.2018.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/30/2018] [Accepted: 05/08/2018] [Indexed: 10/16/2022]
|
82
|
Koh HK, Seo SY, Kim JH, Kim HJ, Chie EK, Kim SK, Kim IH. Disulfiram, a Re-positioned Aldehyde Dehydrogenase Inhibitor, Enhances Radiosensitivity of Human Glioblastoma Cells In Vitro. Cancer Res Treat 2018; 51:696-705. [PMID: 30121967 PMCID: PMC6473295 DOI: 10.4143/crt.2018.249] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/10/2018] [Indexed: 11/21/2022] Open
Abstract
Purpose Glioblastoma, the most common brain tumor in adults, has poor prognosis. The purpose of this study was to determine the effect of disulfiram (DSF), an aldehyde dehydrogenase inhibitor, on in vitro radiosensitivity of glioblastoma cells with different methylation status of O6-methylguanine-DNA methyltransferase (MGMT) promoter and the underlying mechanism of such effect. Materials and Methods Five human glioblastoma cells (U138MG, T98G, U251MG, U87MG, and U373MG) and one normal human astrocyte (NHA) cell were cultured and treated with DSF or 6MV X-rays (0, 2, 4, 6, and 8 Gy). For combined treatment, cells were treated with DSF before irradiation. Surviving fractions fit from cell survival based on colony forming ability. Apoptosis, DNA damage repair, and cell cycle distributionwere assayed bywestern blot for cleaved caspase-3, γH2AX staining, and flow cytometry, respectively. Results DSF induced radiosensitization in most of the glioblastoma cells, especially, in the cells with radioresistance as wildtype unmethylated promoter (MGMT-wt), but did not in normal NHA cell. DSF augmented or induced cleavage of caspase-3 in all cells after irradiation. DSF inhibited repair of radiation-induced DNA damage in MGMT-wt cells, but not in cells with methylated MGMT promoter. DSF abrogated radiation-induced G2/M arrest in T98G and U251MG cells. Conclusion Radiosensitivity of glioblastoma cells were preferentially enhanced by pre-irradiation DSF treatment compared to normal cell, especially radioresistant cells such as MGMT-wt cells. Induction of apoptosis or inhibition of DNA damage repair may underlie DSF-induced radiosensitization. Clinical benefit of combining DSF with radiotherapy should be investigated in the future.
Collapse
Affiliation(s)
- Hyeon Kang Koh
- Department of Radiation Oncology, Konkuk University Medical Center, Seoul, Korea.,Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Yeon Seo
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Jin Ho Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Eui Kyu Chie
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Korea
| | - Il Han Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
83
|
Jayamanne D, Wheeler H, Brazier D, Newey A, Kastelan M, Guo L, Back M. Predicting patterns of failure in temporal lobe GBMs: possible implications on radiotherapy treatment portals. Radiat Oncol 2018; 13:133. [PMID: 30029668 PMCID: PMC6053721 DOI: 10.1186/s13014-018-1078-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/12/2018] [Indexed: 12/25/2022] Open
Abstract
Background Characterise patterns of failure of Temporal Lobe (TL) Glioblastoma (GBM) following treatment with relation to normal temporal lobe anatomy and neural pathways. Methods 335 GBM patients received radiotherapy between 03/2007 and 07/2014, 100 were located in TL. Site of initial tumour and subsequent relapse were subdivided into 5 local TL sites (anterior, lateral, medial, posterior and superior); 5 adjacent regional sites (occipital lobe, inferior frontal lobe, caudate/thalamus/internal/external capsules, fornix/ventricular trigone), and 5 distant failure sites (ventricles, contralateral hemisphere, brainstem, leptomeninges and spine). Extension along major neuroanatomical pathways at initial presentation and at first documented Magnetic Resonance Imaging (MRI) failure were categorised into anterior, superior, medial and posterior pathways. Results Of the 100 patients, 86 had radiological progress with a median survival of 17.3 months. At initial diagnosis, 74% of tumours were linked to one TL site and 94% were confined to the TL. 19% had neural pathway disease at initial pre-treatment MRI. At first recurrence locoregional site failure was 74%. 26% failed within distant sites and 53% patients were noted to have neural pathway involvement. Initial tumour location predicted for local site recurrence (p < 0.0001), regional site recurrence (p = 0.004) and neural pathway recurrence pattern (p = 0.005), but not for distant sites (p = 0.081). Conclusion Most GBMs fail at local or adjacent regional sites. Many of the recurrences occurred in a predictable pattern within a local or regional site, unique to initial TL site with more than half involving neural pathways. Knowledge of tumour infiltration and failure may improve target definition and radiotherapy.
Collapse
Affiliation(s)
- Dasantha Jayamanne
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia. .,Central Coast Cancer Centre, Gosford Hospital, Gosford, NSW, Australia.
| | - Helen Wheeler
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia.,Northern Clinical School, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Sydney Neurooncology Group, Sydney, NSW, Australia
| | - David Brazier
- Department of Radiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Allison Newey
- Department of Radiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Marina Kastelan
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia.,Sydney Neurooncology Group, Sydney, NSW, Australia
| | - Linxin Guo
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Michael Back
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia.,Northern Clinical School, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Central Coast Cancer Centre, Gosford Hospital, Gosford, NSW, Australia.,Sydney Neurooncology Group, Sydney, NSW, Australia
| |
Collapse
|
84
|
Fontanilles M, Marguet F, Alexandru C, Langlois O, Veresezan O, Gilard V, David M, Laquerriere A, Hanzen C, Tennevet I, Di Fiore F, Clatot F. Early platelet variation during concomitant chemo-radiotherapy predicts adjuvant temozolomide-induced thrombocytopenia in newly diagnosed glioblastoma patients. Support Care Cancer 2018; 27:477-484. [PMID: 29978325 DOI: 10.1007/s00520-018-4336-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/26/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Temozolomide (TMZ) is known to induce thrombocytopenia but no early predictive test has yet been clearly established. The aim of the study was to retrospectively identify and validate a threshold of early platelet variation predicting TMZ-induced thrombocytopenia during the TMZ phase in patients treated according to the Stupp protocol for glioblastoma. METHODS A training set was used to analyze variations in platelet count occurring from the first week (W1) to week 6 (W6) during radiotherapy. Our aim was to identify the most relevant platelet decrease associated with TMZ-induced thrombocytopenia ≤ 100 G/L at day 28 during the TMZ phase. The performance of the threshold was confirmed in an independent validation set. RESULTS Overall, 147 patients were included, 85 and 62 in the training and validation sets, respectively. Twenty-seven patients (18%) experienced at least one TMZ-induced thrombocytopenia in the TMZ phase. A platelet decrease at W6 ≥ 35% (∆W6 ≥ 35%) was identified as the best predictive variation with an AUC of 0.83, a sensitivity of 65%, and a specificity of 96%. In the validation set, ∆W6 ≥ 35% platelet variation was identified as an independent marker of TMZ-induced thrombocytopenia during the TMZ phase (OR 15.23 (95% CI 3.5-107.5)) corresponding to sensitivity of 77% (66-87%), specificity of 73% (62-84%), a positive predictive value of 42% (29-54%), and a negative predictive value of 92% (86-99%). CONCLUSION Platelet decrease at W6 ≥ 35% during the RT-TMZ phase is an early and simple predictive marker of clinically relevant TMZ-induced thrombocytopenia during TMZ maintenance.
Collapse
Affiliation(s)
- Maxime Fontanilles
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, F-76031, Rouen, France. .,Department of Medical Oncology, Cancer Centre Henri Becquerel, 1 Rue d'Amiens, 76000, Rouen Cedex, France.
| | - Florent Marguet
- Normandie Univ, UNIROUEN, Inserm U1245, Normandy Centre for Genomic and Personalized Medicine, Department of Pathology, Rouen University Hospital, F-76031, Rouen, France
| | - Cristina Alexandru
- Department of Medical Oncology, Cancer Centre Henri Becquerel, 1 Rue d'Amiens, 76000, Rouen Cedex, France
| | - Olivier Langlois
- Department of Neurosurgery, Rouen University Hospital, F-76031, Rouen, France
| | - Ovidiu Veresezan
- Department of Radiation Oncology and Medical Physics, Cancer Centre Henri Becquerel, F-76000, Rouen, France
| | - Vianney Gilard
- Department of Neurosurgery, Rouen University Hospital, F-76031, Rouen, France
| | - Marion David
- Department of Biopathology, Cancer Centre Henri Becquerel, F-76000, Rouen, France
| | - Annie Laquerriere
- Normandie Univ, UNIROUEN, Inserm U1245, Normandy Centre for Genomic and Personalized Medicine, Department of Pathology, Rouen University Hospital, F-76031, Rouen, France
| | - Chantal Hanzen
- Department of Neurosurgery, Rouen University Hospital, F-76031, Rouen, France
| | - Isabelle Tennevet
- Department of Medical Oncology, Cancer Centre Henri Becquerel, 1 Rue d'Amiens, 76000, Rouen Cedex, France
| | - Frédéric Di Fiore
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, F-76031, Rouen, France.,Department of Medical Oncology, Cancer Centre Henri Becquerel, 1 Rue d'Amiens, 76000, Rouen Cedex, France.,Department of Hepatogastroenterology, Rouen University Hospital, F-76031, Rouen, France
| | - Florian Clatot
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, F-76031, Rouen, France.,Department of Medical Oncology, Cancer Centre Henri Becquerel, 1 Rue d'Amiens, 76000, Rouen Cedex, France
| |
Collapse
|
85
|
Blumenthal DT, Gorlia T, Gilbert MR, Kim MM, Burt Nabors L, Mason WP, Hegi ME, Zhang P, Golfinopoulos V, Perry JR, Hyun Nam D, Erridge SC, Corn BW, Mirimanoff RO, Brown PD, Baumert BG, Mehta MP, van den Bent MJ, Reardon DA, Weller M, Stupp R. Is more better? The impact of extended adjuvant temozolomide in newly diagnosed glioblastoma: a secondary analysis of EORTC and NRG Oncology/RTOG. Neuro Oncol 2018; 19:1119-1126. [PMID: 28371907 DOI: 10.1093/neuonc/nox025] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Radiation with concurrent and adjuvant (6 cycles) temozolomide (TMZ) is the established standard of postsurgical care for newly diagnosed glioblastoma (GBM). This regimen has been adopted with variations, including extending TMZ beyond 6 cycles. The optimal duration of maintenance therapy remains controversial. Methods We performed pooled analysis of individual patient data from 4 randomized trials for newly diagnosed GBM. All patients who were progression free 28 days after cycle 6 were included. The decision to continue TMZ was per local practice and standards, and at the discretion of the treating physician. Patients were grouped into those treated with 6 cycles and those who continued beyond 6 cycles. Progression-free and overall survival were compared, adjusted by age, performance status, resection extent, and MGMT methylation. Results A total of 2214 GBM patients were included in the 4 trials. Of these, 624 qualified for analysis 291 continued maintenance TMZ until progression or up to 12 cycles, while 333 discontinued TMZ after 6 cycles. Adjusted for prognostic factors, treatment with more than 6 cycles of TMZ was associated with a somewhat improved progression-free survival (hazard ratio [HR] 0.80 [0.65-0.98], P = .03), in particular for patients with methylated MGMT (n = 342, HR 0.65 [0.50-0.85], P < .01). However, overall survival was not affected by the number of TMZ cycles (HR = 0.92 [0.71-1.19], P = .52), including the MGMT methylated subgroup (HR = 0.89 [0.63-1.26], P = .51). Conclusions Continuing TMZ beyond 6 cycles was not shown to increase overall survival for newly diagnosed GBM.
Collapse
Affiliation(s)
- Deborah T Blumenthal
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Thierry Gorlia
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Mark R Gilbert
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Michelle M Kim
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - L Burt Nabors
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Warren P Mason
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Monika E Hegi
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Peixin Zhang
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Vassilis Golfinopoulos
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - James R Perry
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Do Hyun Nam
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Sara C Erridge
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Benjamin W Corn
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - René O Mirimanoff
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Paul D Brown
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Brigitta G Baumert
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Minesh P Mehta
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Martin J van den Bent
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - David A Reardon
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Michael Weller
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Roger Stupp
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| |
Collapse
|
86
|
Rapp M, Grauer OM, Kamp M, Sevens N, Zotz N, Sabel M, Sorg RV. A randomized controlled phase II trial of vaccination with lysate-loaded, mature dendritic cells integrated into standard radiochemotherapy of newly diagnosed glioblastoma (GlioVax): study protocol for a randomized controlled trial. Trials 2018; 19:293. [PMID: 29801515 PMCID: PMC5970474 DOI: 10.1186/s13063-018-2659-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/02/2018] [Indexed: 01/06/2023] Open
Abstract
Background Despite the combination of surgical resection, radio- and chemotherapy, median survival of glioblastoma multiforme (GBM) patients only slightly increased in the last years. Disease recurrence is definite with no effective therapy existing after tumor removal. Dendritic cell (DC) vaccination is a promising active immunotherapeutic approach. There is clear evidence that it is feasible, results in immunological anti-tumoral responses, and appears to be beneficial for survival and quality of life of GBM patients. Moreover, combining it with the standard therapy of GBM may allow exploiting synergies between the treatment modalities. In this randomized controlled trial, we seek to confirm these promising initial results. Methods One hundred and thirty-six newly diagnosed, isocitrate dehydrogenase wildtype GBM patients will be randomly allocated (1:1 ratio, stratified by O6-methylguanine-DNA-methyltransferase promotor methylation status) after near-complete resection in a multicenter, prospective phase II trial into two groups: (1) patients receiving the current therapeutic “gold standard” of radio/temozolomide chemotherapy and (2) patients receiving DC vaccination as an add-on to the standard therapy. A recruitment period of 30 months is anticipated; follow-up will be 2 years. The primary objective of the study is to compare overall survival (OS) between the two groups. Secondary objectives are comparing progression-free survival (PFS) and 6-, 12- and 24-month OS and PFS rates, the safety profile, overall and neurological performance and quality of life. Discussion Until now, close to 500 GBM patients have been treated with DC vaccination in clinical trials or on a compassionate-use basis. Results have been encouraging, but cannot provide robust evidence of clinical efficacy because studies have been non-controlled or patient numbers have been low. Therefore, a prospective, randomized phase II trial with a sufficiently large number of patients is now mandatory for clear evidence regarding the impact of DC vaccination on PFS and OS in GBM. Trial registration Protocol code: GlioVax, date of registration: 17. February 2017. Trial identifier: EudraCT-Number 2017–000304-14. German Registry for Clinical Studies, ID: DRKS00013248 (approved primary register in the WHO network) and at ClinicalTrials.gov, ID: NCT03395587. Registered on 11 March 2017. Electronic supplementary material The online version of this article (10.1186/s13063-018-2659-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marion Rapp
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany. .,Department of Neurosurgery, Heinrich Heine University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - Oliver M Grauer
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Marcel Kamp
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Natalie Sevens
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Nikola Zotz
- Coordination Center for Clinical Trials, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Michael Sabel
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Rüdiger V Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| |
Collapse
|
87
|
Alterations in Cell Motility, Proliferation, and Metabolism in Novel Models of Acquired Temozolomide Resistant Glioblastoma. Sci Rep 2018; 8:7222. [PMID: 29740146 PMCID: PMC5940876 DOI: 10.1038/s41598-018-25588-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive and incurable tumor of the brain with limited treatment options. Current first-line standard of care is the DNA alkylating agent temozolomide (TMZ), but this treatment strategy adds only ~4 months to median survival due to the rapid development of resistance. While some mechanisms of TMZ resistance have been identified, they are not fully understood. There are few effective strategies to manage therapy resistant GBM, and we lack diverse preclinical models of acquired TMZ resistance in which to test therapeutic strategies on TMZ resistant GBM. In this study, we create and characterize two new GBM cell lines resistant to TMZ in vitro, based on the 8MGBA and 42MGBA cell lines. Analysis of the TMZ resistant (TMZres) variants in conjunction with their parental, sensitive cell lines shows that acquisition of TMZ resistance is accompanied by broad phenotypic changes, including increased proliferation, migration, chromosomal aberrations, and secretion of cytosolic lipids. Importantly, each TMZ resistant model captures a different facet of the “go” (8MGBA-TMZres) or “grow” (42MGBA-TMZres) hypothesis of GBM behavior. These in vitro model systems will be important additions to the available tools for investigators seeking to define molecular mechanisms of acquired TMZ resistance.
Collapse
|
88
|
Li H, Wu JG, Zhang HW, Wang W, Zhang YX, Zhang JN. Inhibition of Proliferation, Invasion and Migration in U-251 MG Glioblastoma Cell Line by Gedunin. INT J PHARMACOL 2018. [DOI: 10.3923/ijp.2018.522.527] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
89
|
Prognostic value of the Glasgow Prognostic Score for glioblastoma multiforme patients treated with radiotherapy and temozolomide. J Neurooncol 2018; 139:411-419. [PMID: 29696530 DOI: 10.1007/s11060-018-2879-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/22/2018] [Indexed: 01/01/2023]
Abstract
INTRODUCTION To evaluate the prognostic value of the Glasgow Prognostic Score (GPS), the combination of C-reactive protein (CRP) and albumin, in glioblastoma multiforme (GBM) patients treated with radiotherapy (RT) and concurrent plus adjuvant temozolomide (GPS). METHODS Data of newly diagnosed GBM patients treated with partial brain RT and concurrent and adjuvant TMZ were retrospectively analyzed. The patients were grouped into three according to the GPS criteria: GPS-0: CRP < 10 mg/L and albumin > 35 g/L; GPS-1: CRP < 10 mg/L and albumin < 35 g/L or CRP > 10 mg/L and albumin > 35 g/L; and GPS-2: CRP > 10 mg/L and albumin < 35 g/L. Primary end-point was the association between the GPS groups and the overall survival (OS) outcomes. RESULTS A total of 142 patients were analyzed (median age: 58 years, 66.2% male). There were 64 (45.1%), 40 (28.2%), and 38 (26.7%) patients in GPS-0, GPS-1, and GPS-2 groups, respectively. At median 15.7 months follow-up, the respective median and 5-year OS rates for the whole cohort were 16.2 months (95% CI 12.7-19.7) and 9.5%. In multivariate analyses GPS grouping emerged independently associated with the median OS (P < 0.001) in addition to the extent of surgery (P = 0.032), Karnofsky performance status (P = 0.009), and the Radiation Therapy Oncology Group recursive partitioning analysis (RTOG RPA) classification (P < 0.001). The GPS grouping and the RTOG RPA classification were found to be strongly correlated in prognostic stratification of GBM patients (correlation coefficient: 0.42; P < 0.001). CONCLUSIONS The GPS appeared to be useful in prognostic stratification of GBM patients into three groups with significantly different survival durations resembling the RTOG RPA classification.
Collapse
|
90
|
Huang B, Dolecek TA, Chen Q, Garcia CR, Pittman T, Villano JL. Characteristics and survival outcomes associated with the lack of radiation in the treatment of glioblastoma. Med Oncol 2018; 35:74. [PMID: 29667068 DOI: 10.1007/s12032-018-1134-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Radiation increases survival in glioblastoma (GBM); however, 30% do not receive this treatment. We sought to identify characteristics associated with not receiving radiation and the impact on outcomes. We analyzed the Surveillance, Epidemiology, and End Results program (SEER) 18 registries 2000-2013 research database on 30,479 GBM cases that were aged 20 years and older. In total, 21,179 received radiation as first course of therapy, while 8218 did not with 5178 (63%) being 65 years and older. Early decisions on surgery often predicted radiation therapy with 61% having only a biopsy or no surgery at diagnosis. Radiation use as upfront therapy has slowly increased over time at a rate of 0.4% per year; still 25% did not receive radiation in 2013. Cases treated with radiation were more likely to be younger, underwent surgery, lived in a metropolitan area, had higher socioeconomic status, and were in a couple-based relationship. An increased survival in GBM was associated with the use of upfront radiation along with younger age, being of race other than white, undergoing surgery, and a more recent diagnosis. Not receiving radiation therapy adversely affects survival. A trend toward an increased use of radiation was observed although many young adults still do not receive this treatment. Decreased usage of radiation in the elderly and in biopsy-only surgeries was anticipated, but race, gender, and poverty were also statistically significant. Clinicians should be aware of this underutilization, and an increased usage of radiation should improve outcomes for glioblastoma.
Collapse
Affiliation(s)
- Bin Huang
- Division of Cancer Biostatistics, College of Public Health, University of Kentucky, 2365 Harrodsburg Road Suite A230, Lexington, KY, 40504, USA.,Markey Cancer Center, University of Kentucky, 800 Rose St., Lexington, KY, 40536-0093, USA
| | - Therese A Dolecek
- Division of Epidemiology and Biostatistics and Institute for Health Research and Policy, School of Public Health, University of Illinois at Chicago, 809 South Marshfield Avenue, MB 502, M/C 551, Chicago, IL, 60612-7205, USA
| | - Quan Chen
- Markey Cancer Center, University of Kentucky, 800 Rose St., Lexington, KY, 40536-0093, USA
| | - Catherine R Garcia
- Markey Cancer Center, University of Kentucky, 800 Rose St., Lexington, KY, 40536-0093, USA
| | - Thomas Pittman
- Department of Neurosurgery, University of Kentucky, MS105 Medical Science Building, Lexington, KY, 40536-0298, USA.,Markey Cancer Center, University of Kentucky, 800 Rose St., Lexington, KY, 40536-0093, USA
| | - John L Villano
- Department of Neurology, University of Kentucky, Lexington, KY, USA. .,Department of Medicine, University of Kentucky, Lexington, KY, USA. .,Department of Neurosurgery, University of Kentucky, MS105 Medical Science Building, Lexington, KY, 40536-0298, USA. .,Markey Cancer Center, University of Kentucky, 800 Rose St., Lexington, KY, 40536-0093, USA.
| |
Collapse
|
91
|
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
|
92
|
Lecler A, Charbonneau F, Psimaras D, Metten MA, Gueguen A, Hoang Xuan K, Feuvret L, Savatovsky J. Remote brain microhaemorrhages may predict haematoma in glioma patients treated with radiation therapy. Eur Radiol 2018; 28:4324-4333. [PMID: 29651771 DOI: 10.1007/s00330-018-5356-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/03/2018] [Accepted: 01/26/2018] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To evaluate the prevalence of cerebral remote microhaemorrhages (RMH) and remote haematomas (RH) using magnetic resonance susceptibility-weighted imaging (SWI) among patients treated for gliomas during follow-up. METHODS We conducted a retrospective single centre longitudinal study on 58 consecutive patients treated for gliomas from January 2009 through December 2010. Our institutional review board approved this study. We evaluated the presence and number of RMH and RH found outside the brain tumour on follow-up MR imaging. We performed univariate and bivariate analyses to identify predictors for RMH and RH and Kaplan-Meier survival analysis techniques. RESULTS Twenty-five (43%) and four patients (7%) developed at least one RMH or RH, respectively, during follow-up. The risk was significantly higher for patients who received radiation therapy (49% and 8% versus 0%) (p = 0.02). The risk of developing RH was significantly higher in patients with at least one RMH and a high burden of RMH. The mean age of those presenting with at least one RMH or RH was significantly lower. CONCLUSIONS RMH were common in adult survivors of gliomas who received radiation therapy and may predict the onset of RH during follow-up, mainly in younger patients. KEY POINTS • Brain RMH and RH are significantly more likely to occur after RT. • RMH occur in almost half of the patients treated with RT. • RMH and RH are significantly more frequent in younger patients. • RH occur only in patients with RMH.
Collapse
Affiliation(s)
- Augustin Lecler
- Department of Radiology, Fondation Ophtalmologique Adolphe de Rothschild, 25 rue Manin, 75019, Paris, France.
| | - Frédérique Charbonneau
- Department of Radiology, Fondation Ophtalmologique Adolphe de Rothschild, 25 rue Manin, 75019, Paris, France
| | - Dimitri Psimaras
- Department of Neurology, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, Paris, France
| | - Marie-Astrid Metten
- Clinical Research Unit, Fondation Ophtalmologique Adolphe de Rothschild, Paris, France
| | - Antoine Gueguen
- Department of Neurology, Fondation Ophtalmologique Adolphe de Rothschild, Paris, France
| | - Khe Hoang Xuan
- Department of Neurooncology, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, Paris, France
| | - Loic Feuvret
- Department of Radiotherapy, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, Paris, France
| | - Julien Savatovsky
- Department of Radiology, Fondation Ophtalmologique Adolphe de Rothschild, 25 rue Manin, 75019, Paris, France.,Imagerie Medicale Paris 13, Paris, France
| |
Collapse
|
93
|
Predicting the cell death responsiveness and sensitization of glioma cells to TRAIL and temozolomide. Oncotarget 2018; 7:61295-61311. [PMID: 27494880 PMCID: PMC5308652 DOI: 10.18632/oncotarget.10973] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 07/18/2016] [Indexed: 12/28/2022] Open
Abstract
Genotoxic chemotherapy with temozolomide (TMZ) is a mainstay of treatment for glioblastoma (GBM); however, at best, TMZ provides only modest survival benefit to a subset of patients. Recent insight into the heterogeneous nature of GBM suggests a more personalized approach to treatment may be necessary to overcome cancer drug resistance and improve patient care. These include novel therapies that can be used both alone and with TMZ to selectively reactivate apoptosis within malignant cells. For this approach to work, reliable molecular signatures that can accurately predict treatment responsiveness need to be identified first. Here, we describe the first proof-of-principle study that merges quantitative protein-based analysis of apoptosis signaling networks with data- and knowledge-driven mathematical systems modeling to predict treatment responsiveness of GBM cell lines to various apoptosis-inducing stimuli. These include monotherapies with TMZ and TRAIL, which activate the intrinsic and extrinsic apoptosis pathways, respectively, as well as combination therapies of TMZ+TRAIL. We also successfully employed this approach to predict whether individual GBM cell lines could be sensitized to TMZ or TRAIL via the selective targeting of Bcl-2/Bcl-xL proteins with ABT-737. Our findings suggest that systems biology-based approaches could assist in personalizing treatment decisions in GBM to optimize cell death induction.
Collapse
|
94
|
Jackson WC, Tsien CI, Junck L, Leung D, Hervey-Jumper S, Orringer D, Heth J, Wahl DR, Spratt DE, Cao Y, Lawrence TS, Kim MM. Standard dose and dose-escalated radiation therapy are associated with favorable survival in select elderly patients with newly diagnosed glioblastoma. J Neurooncol 2018; 138:155-162. [DOI: 10.1007/s11060-018-2782-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/29/2018] [Indexed: 11/24/2022]
|
95
|
Raverot G, Burman P, McCormack A, Heaney A, Petersenn S, Popovic V, Trouillas J, Dekkers OM. European Society of Endocrinology Clinical Practice Guidelines for the management of aggressive pituitary tumours and carcinomas. Eur J Endocrinol 2018; 178:G1-G24. [PMID: 29046323 DOI: 10.1530/eje-17-0796] [Citation(s) in RCA: 332] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/12/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Pituitary tumours are common and easily treated by surgery or medical treatment in most cases. However, a small subset of pituitary tumours does not respond to standard medical treatment and presents with multiple local recurrences (aggressive pituitary tumours) and in rare occasion with metastases (pituitary carcinoma). The present European Society of Endocrinology (ESE) guideline aims to provide clinical guidance on diagnosis, treatment and follow-up in aggressive pituitary tumours and carcinomas. METHODS We decided upfront, while acknowledging that literature on aggressive pituitary tumours and carcinomas is scarce, to systematically review the literature according to the GRADE (Grading of Recommendations Assessment, Development and Evaluation) system. The review focused primarily on first- and second-line treatment in aggressive pituitary tumours and carcinomas. We included 14 single-arm cohort studies (total number of patients = 116) most on temozolomide treatment (n = 11 studies, total number of patients = 106). A positive treatment effect was seen in 47% (95% CI: 36-58%) of temozolomide treated. Data from the recently performed ESE survey on aggressive pituitary tumours and carcinomas (165 patients) were also used as backbone for the guideline. SELECTED RECOMMENDATION: (i) Patients with aggressive pituitary tumours should be managed by a multidisciplinary expert team. (ii) Histopathological analyses including pituitary hormones and proliferative markers are needed for correct tumour classification. (iii) Temozolomide monotherapy is the first-line chemotherapy for aggressive pituitary tumours and pituitary carcinomas after failure of standard therapies; treatment evaluation after 3 cycles allows identification of responder and non-responder patients. (iv) In patients responding to first-line temozolomide, we suggest continuing treatment for at least 6 months in total. Furthermore, the guideline offers recommendations for patients who recurred after temozolomide treatment, for those who did not respond to temozolomide and for patients with systemic metastasis.
Collapse
Affiliation(s)
- Gerald Raverot
- Fédération d'Endocrinologie, Centre de Référence des Maladies Rares Hypophysaires HYPO, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Faculté de Médecine Lyon Est, Université Lyon 1, Lyon, France
- INSERM U1052, CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France
| | - Pia Burman
- Department of Endocrinology, Skane University Hospital Malmö, University of Lund, Lund, Sweden
| | - Ann McCormack
- Garvan Institute, Sydney, Australia
- Department of Endocrinology, St Vincent's Hospital, University of New South Wales, Sydney, Australia
| | - Anthony Heaney
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | - Vera Popovic
- Medical Faculty, University Belgrade, Belgrade, Serbia
| | - Jacqueline Trouillas
- Faculté de Médecine Lyon Est, Université Lyon 1, Lyon, France
- Centre de Pathologie et de Biologie Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
| | - Olaf M Dekkers
- Departments of Internal Medicine (Section Endocrinology) & Clinical Epidemiology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
96
|
Beez T, Burgula S, Kamp M, Rapp M, Steiger HJ, Sabel M. Symptomatic communicating hydrocephalus in a contemporary cohort of high grade glioma patients. Br J Neurosurg 2017; 32:68-72. [PMID: 29160137 DOI: 10.1080/02688697.2017.1380780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION High grade glioma (HGG) treatment has seen a paradigm shift with intensified regimes, but hospitalisation burden is partially attributed to side effects of therapy. Symptomatic communicating hydrocephalus (HC) is a potential complication. Aim of this study was to investigate its incidence, risk factors and treatment in a contemporary cohort. METHODS We performed a retrospective review of HGG patients. Patients with symptomatic communicating HC were identified and demographic, tumour and treatment variables extracted from hospital notes. Descriptive statistics were performed and odds ratios (OR) with 95% confidence intervals (CI) calculated. RESULTS From a cohort of 278 eligible HGG patients, 8 (3%) were diagnosed with symptomatic communicating HC. In the subgroup of patients with intraoperative opening of the ventricular system during previous surgery (N = 66), hydrocephalus developed in 10.6% of cases (N = 7). Symptoms were lethargy (N = 5), headache (N = 3), confusion (N = 3), gait disturbance (N = 3) and urinary incontinence (N = 1); three patients presented resembling normal pressure hydrocephalus. A higher rate of intraoperative opening of the ventricles was found in the HC group (p = .0002); all other variables were equally distributed. The increased odds for developing HC for patients with intraoperative opening of the ventricles reached statistical significance (OR = 25.0339, 95% CI = 3.0196-207.5449, p = .0028), whereas previous radiotherapy only increased odds by tendency. Hydrocephalic patients were treated with ventriculoperitoneal shunts and all but one patient improved. One patient had a complication attributed to the shunt procedure The median overall survival after shunting was 4 months. CONCLUSIONS Symptomatic communicating hydrocephalus occurred in 3% of HGG patients and in 10.6% in the subgroup with previous intraoperative opening of the ventricles. Treatment with a ventriculoperitoneal shunt is effective and has a low complication rate.
Collapse
Affiliation(s)
- Thomas Beez
- a Department of Neurosurgery, Medical Faculty , Heinrich-Heine-University , Düsseldorf , Germany
| | - Sven Burgula
- a Department of Neurosurgery, Medical Faculty , Heinrich-Heine-University , Düsseldorf , Germany
| | - Marcel Kamp
- a Department of Neurosurgery, Medical Faculty , Heinrich-Heine-University , Düsseldorf , Germany
| | - Marion Rapp
- a Department of Neurosurgery, Medical Faculty , Heinrich-Heine-University , Düsseldorf , Germany
| | - Hans-Jakob Steiger
- a Department of Neurosurgery, Medical Faculty , Heinrich-Heine-University , Düsseldorf , Germany
| | - Michael Sabel
- a Department of Neurosurgery, Medical Faculty , Heinrich-Heine-University , Düsseldorf , Germany
| |
Collapse
|
97
|
Prognostic factors for survival in adult patients with recurrent glioblastoma: a decision-tree-based model. J Neurooncol 2017; 136:565-576. [PMID: 29159777 DOI: 10.1007/s11060-017-2685-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/11/2017] [Indexed: 01/30/2023]
Abstract
We assessed prognostic factors in relation to OS from progression in recurrent glioblastomas. Retrospective multicentric study enrolling 407 (training set) and 370 (external validation set) adult patients with a recurrent supratentorial glioblastoma treated by surgical resection and standard combined chemoradiotherapy as first-line treatment. Four complementary multivariate prognostic models were evaluated: Cox proportional hazards regression modeling, single-tree recursive partitioning, random survival forest, conditional random forest. Median overall survival from progression was 7.6 months (mean, 10.1; range, 0-86) and 8.0 months (mean, 8.5; range, 0-56) in the training and validation sets, respectively (p = 0.900). Using the Cox model in the training set, independent predictors of poorer overall survival from progression included increasing age at histopathological diagnosis (aHR, 1.47; 95% CI [1.03-2.08]; p = 0.032), RTOG-RPA V-VI classes (aHR, 1.38; 95% CI [1.11-1.73]; p = 0.004), decreasing KPS at progression (aHR, 3.46; 95% CI [2.10-5.72]; p < 0.001), while independent predictors of longer overall survival from progression included surgical resection (aHR, 0.57; 95% CI [0.44-0.73]; p < 0.001) and chemotherapy (aHR, 0.41; 95% CI [0.31-0.55]; p < 0.001). Single-tree recursive partitioning identified KPS at progression, surgical resection at progression, chemotherapy at progression, and RTOG-RPA class at histopathological diagnosis, as main survival predictors in the training set, yielding four risk categories highly predictive of overall survival from progression both in training (p < 0.0001) and validation (p < 0.0001) sets. Both random forest approaches identified KPS at progression as the most important survival predictor. Age, KPS at progression, RTOG-RPA classes, surgical resection at progression and chemotherapy at progression are prognostic for survival in recurrent glioblastomas and should inform the treatment decisions.
Collapse
|
98
|
Solid Lipid Curcumin Particles Induce More DNA Fragmentation and Cell Death in Cultured Human Glioblastoma Cells than Does Natural Curcumin. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9656719. [PMID: 29359011 PMCID: PMC5735327 DOI: 10.1155/2017/9656719] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/25/2017] [Accepted: 10/02/2017] [Indexed: 12/17/2022]
Abstract
Despite recent advancements in cancer therapies, glioblastoma multiforme (GBM) remains largely incurable. Curcumin (Cur), a natural polyphenol, has potent anticancer effects against several malignancies, including metastatic brain tumors. However, its limited bioavailability reduces its efficiency for treating GBM. Recently, we have shown that solid lipid Cur particles (SLCPs) have greater bioavailability and brain tissue penetration. The present study compares the efficiency of cell death by Cur and/or SLCPs in cultured GBM cells derived from human (U-87MG) and mouse (GL261) tissues. Several cell viability and cell death assays and marker proteins (MTT assay, annexin-V staining, TUNEL staining, comet assay, DNA gel electrophoresis, and Western blot) were investigated following the treatment of Cur and/or SLCP (25 μM) for 24–72 h. Relative to Cur, the use of SLCP increased cell death and DNA fragmentation, produced longer DNA tails, and induced more fragmented nuclear lobes. In addition, cultured GBM cells had increased levels of caspase-3, Bax, and p53, with decreases in Bcl2, c-Myc, and both total Akt, as well as phosphorylated Akt, when SLCP, rather Cur, was used. Our in vitro work suggests that the use of SLCP may be a promising strategy for reversing or preventing GBM growth, as compared to using Cur.
Collapse
|
99
|
Liang HKT, Chen WY, Lai SF, Su MY, You SL, Chen LH, Tseng HM, Chen CM, Kuo SH, Tseng WYI. The extent of edema and tumor synchronous invasion into the subventricular zone and corpus callosum classify outcomes and radiotherapy strategies of glioblastomas. Radiother Oncol 2017; 125:248-257. [PMID: 29056290 DOI: 10.1016/j.radonc.2017.09.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 08/10/2017] [Accepted: 09/23/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND PURPOSE Irradiating glioblastoma preoperative edema (PE) remains controversial. We investigated the associations between tumors' PE extent with invasion into synchronous subventricular zone and corpus callosum (sSVZCC) and treatment outcomes to provide the clinical evidence for radiotherapy decision-making. MATERIAL AND METHODS Extensive PE (EPE) was defined as PE extending ≥2 cm from the tumor edge and extensive progressive disease (EPD) as tumors spreading ≥2 cm from the preoperative tumor edge along PE. The survival and progression patterns were analyzed according to EPE and sSVZCC invasion. RESULTS In total, 136 patients were followed for a median of 74.9 (range, 47.6-102.1) months. The median overall survival and progression-free survival were 19.7 versus 28.6 months (p = 0.005) and 11.0 versus 17.4 months (p = 0.011) in patients with EPE+ versus EPE-, and were 18.7 versus 25.4 months (p = 0.021) and 10.7 versus 14.6 months (p = 0.020) in those with sSVZCC+ versus sSVZCC-. The EPD rates for tumors with EPE-/sSVZCC-, EPE-/sSVZCC+, EPE+/sSVZCC-, and EPE+/sSVZCC+ were 2.8%, 7.1%, 37.0%, and 71.9%, respectively. In EPE+/sSVZCC+, tumor migration was associated with the PE extending along the corpus callosum (77.8%) and subventricular zone (50.0%). CONCLUSIONS Our results support the need for developing individualized irradiation strategies for glioblastomas according to EPE and sSVZCC.
Collapse
Affiliation(s)
- Hsiang-Kuang Tony Liang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan; Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Radiation Science and Proton Therapy Center, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Neurology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wan-Yu Chen
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Radiation Science and Proton Therapy Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shih-Fan Lai
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan; Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Radiation Science and Proton Therapy Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Mao-Yuan Su
- Department of Medical Imaging, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - San-Lin You
- School of Medicine, College of Medicine, and Big Data Research Center, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Liang-Hsin Chen
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan; Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Radiation Science and Proton Therapy Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ham-Min Tseng
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chung-Ming Chen
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Sung-Hsin Kuo
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan; Radiation Science and Proton Therapy Center, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Yih Isaac Tseng
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
100
|
Bettega D, Calzolari P, Ciocca M, Facoetti A, Lafiandra M, Marchesini R, Molinelli S, Pignoli E, Vischioni B. Combining proton or photon irradiation with epothilone B. An
in vitro
study of cytotoxicity in human cancer cells. Biomed Phys Eng Express 2017. [DOI: 10.1088/2057-1976/aa818f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|