51
|
Wang Z, Xie F, Wu Y, Wang L, Bai Y, Long J, Wang X. Differential genes and scoring criteria among immunogenomic clusters of lower-grade gliomas. Int Immunopharmacol 2021; 101:108376. [PMID: 34815191 DOI: 10.1016/j.intimp.2021.108376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/28/2021] [Accepted: 11/11/2021] [Indexed: 01/06/2023]
Abstract
High incidence of recurrency had been a significant threat among glioma patients. Moreover, the performance of traditional therapies among recurrent gliomas was far from satisfying. Advances in the tumor microenvironment (TME) and immune responses on the brain inspired immunotherapy researches. Nevertheless, verification of classic PD-1/PD-L1 inhibitors failed in phase III clinical trials. Additional gene targets were required for future studies among glioma patients. Immune cell infiltration (ICI) scores, defined based on multiple prognostic genes, were proved as the marker for the sensitivity of immunotherapies in many tumors. However, relevant results were not reported in gliomas. In the study, a retrospective cohort of 495 patients was classified into two ICI score subgroups. High ICI scores were closely related to high tumor mutation burden (TMB) values, indicating a high instability of genes. Furthermore, ICI scores were proved as reliable prognostic predictors. And a predictive model was built based on the ICI scores and multiple clinical features. The model showed its superiority through both internal validation and external validation. The ICI scores and the predictive model showed significant clinical values through decision curve analysis (DCA) since high ICI scores were related to high sensitivity for treatment. The prognostic immune-related gene list provided targets for immunotherapy researches.
Collapse
Affiliation(s)
- Zhile Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fucun Xie
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yijun Wu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Li Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yi Bai
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Junyu Long
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiang Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
52
|
An overview of current therapeutic strategies for glioblastoma and the role of CD73 as an alternative curative approach. Clin Transl Oncol 2021; 24:742-756. [PMID: 34792724 DOI: 10.1007/s12094-021-02732-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022]
Abstract
Glioblastoma multiforme (GBM) is a complicated and heterogeneous brain tumor with short-term survival outcomes. Commercial therapies are not practical due to cell infiltration capacity, high proliferative rate, and blood-brain barrier. In this context, recognition of the molecular mechanism of tumor progression might help the development of new cancer therapeutics. Recently, more evidence has supported CD73 and downstream adenosine A2A/A2B receptor signaling playing a crucial role in glioblastoma pathogenesis; therefore, targeting CD73 in murine tumor models can reduce tumor development. CD73 is an ecto-enzyme inducing tumor metastasis, angiogenesis, and immune escape via the production of extracellular adenosine in the tumor microenvironment. In this review, we provided information about clinical characteristics as well as the therapeutic management of glioblastoma. Then, we focused on newly available experimental evidence distinguishing between the essential role of CD73 on this tumor growth and a new method for the treatment of GBM patients.
Collapse
|
53
|
Qu S, Qiu O, Hu Z. The prognostic factors and nomogram for patients with high-grade gliomas. FUNDAMENTAL RESEARCH 2021. [DOI: 10.1016/j.fmre.2021.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
54
|
Lavogina D, Laasfeld T, Vardja M, Lust H, Jaal J. Viability fingerprint of glioblastoma cell lines: roles of mitotic, proliferative, and epigenetic targets. Sci Rep 2021; 11:20338. [PMID: 34645858 PMCID: PMC8514540 DOI: 10.1038/s41598-021-99630-0] [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: 08/03/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023] Open
Abstract
Despite the use of multimodal treatment combinations, the prognosis of glioblastoma (GB) is still poor. To prevent rapid tumor recurrence, targeted strategies for the treatment of GB are widely sought. Here, we compared the efficacy of focused modulation of a set of signaling pathways in two GB cell lines, U-251 MG and T98-G, using a panel of thirteen compounds targeting cell cycle progression, proliferation, epigenetic modifications, and DNA repair mechanism. In parallel, we tested combinations of these compounds with temozolomide and lomustine, the standard chemotherapy agents used in GB treatment. Two major trends were found: within individual compounds, the lowest IC50 values were exhibited by the Aurora kinase inhibitors, whereas in the case of mixtures, the addition of DNA methyltransferase 1 inhibitor azacytidine to lomustine proved the most beneficial. The efficacy of cell cycle-targeting compounds was further augmented by combination with radiation therapy using two different treatment regimes. The potency of azacytidine and lomustine mixtures was validated using a unique assay pipeline that utilizes automated imaging and machine learning-based data analysis algorithm for assessment of cell number and DNA damage extent. Based on our results, the combination of azacytidine and lomustine should be tested in GB clinical trials.
Collapse
Affiliation(s)
- Darja Lavogina
- grid.10939.320000 0001 0943 7661Institute of Clinical Medicine, University of Tartu, L. Puusepa 8, 50406 Tartu, Estonia ,grid.10939.320000 0001 0943 7661Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Tõnis Laasfeld
- grid.10939.320000 0001 0943 7661Institute of Chemistry, University of Tartu, Tartu, Estonia ,grid.10939.320000 0001 0943 7661Department of Computer Science, University of Tartu, Tartu, Estonia
| | - Markus Vardja
- grid.412269.a0000 0001 0585 7044Department of Radiotherapy and Oncological Therapy, Tartu University Hospital, Tartu, Estonia
| | - Helen Lust
- grid.10939.320000 0001 0943 7661Institute of Clinical Medicine, University of Tartu, L. Puusepa 8, 50406 Tartu, Estonia
| | - Jana Jaal
- grid.10939.320000 0001 0943 7661Institute of Clinical Medicine, University of Tartu, L. Puusepa 8, 50406 Tartu, Estonia ,grid.412269.a0000 0001 0585 7044Department of Radiotherapy and Oncological Therapy, Tartu University Hospital, Tartu, Estonia
| |
Collapse
|
55
|
Adegboyega G, Ozair A, Kanmounye US, Bandyopadhyay S, Vaqas B. Letter: Is the Stupp Protocol an Expensive and Unsustainable Standard of Care for Glioblastoma in Low- and Middle-Income Country Settings? A Call to Action! Neurosurgery 2021; 89:E249-E251. [PMID: 34318884 DOI: 10.1093/neuros/nyab273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Gideon Adegboyega
- Barts and The London School of Medicine and Dentistry Queen Mary University of London London, UK
- Research Department Association of Future African Neurosurgeons Yaounde, Cameroon
| | - Ahmad Ozair
- Faculty of Medicine King George's Medical University Lucknow, India
| | | | - Soham Bandyopadhyay
- Oxford University Global Surgery Group Nuffield Department of Surgical Sciences University of Oxford Oxford, UK
| | - Babar Vaqas
- Department of Neurosurgery Queens Hospital Romford, UK
- Imperial College London London, UK
| |
Collapse
|
56
|
Mizumoto M, Liang HK, Oshiro Y, Matsuda M, Kohzuki H, Iizumi T, Numajiri H, Nakai K, Okumura T, Ishikawa E, Sakurai H. Radiation Therapy for Grade 3 Gliomas: Correlation of MRI Findings With Prognosis. Cureus 2021; 13:e16887. [PMID: 34513462 PMCID: PMC8416380 DOI: 10.7759/cureus.16887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 11/05/2022] Open
Abstract
Background and objective Postoperative radiotherapy is usually indicated for both grade 3 glioma and grade 4 glioblastoma. However, the treatment results and tumor features of grade 3 glioma clearly differ from those of glioblastoma. There is limited information on outcomes and tumor progression for grade 3 glioma. In this study, we evaluate the result of postoperative radiotherapy for grade 3 glioma and focus on the correlation of MRI findings with prognosis. Methods In this study, 99 of 110 patients with grade 3 glioma who received postoperative radiotherapy and were followed up for more than one year were retrospectively analyzed. The total irradiation dose was 60.0 Gy in 30 fractions, and daily temozolomide or two cycles of nimustine (ACNU) was concurrently administered during radiotherapy. The median follow-up period was 46 months (range: 2-151 months). Results In multivariate analysis, pathology [anaplastic oligodendroglioma (AO) vs. anaplastic astrocytoma (AA)], the status of surgical resection (biopsy vs. partial resection or more), and contrast enhancement (enhanced by MRI image or not) were significant factors for overall survival (OS). The five-year OS for AO vs. AA cases were 76.8% vs. 46.1%, total to partial resection vs. biopsy cases were 72.7% vs. 21.0%, and non-enhanced vs. enhanced cases were 82.5% vs. 45.6%, respectively. In multivariate analysis, the status of surgical resection and longer extension of preoperative edema (PE) were significant factors for progression-free survival (PFS). The five-year PFS for the total to partial resection vs. biopsy cases were 52.9% vs. 10.7%, and non-extensive PE vs. extensive PE (EPE) cases were 62.2% vs. 19.1%, respectively. Conclusion Our results suggest that a contrast-enhanced tumor on MRI and a longer PE may also be significantly associated with OS and PFS among grade 3 glioma patients.
Collapse
Affiliation(s)
- Masashi Mizumoto
- Radiation Oncology, University of Tsukuba Hospital, Tsukuba, JPN
| | - Hsiang-Kuang Liang
- Department of Biomedical Engineering, National Taiwan University, Taipei, TWN.,Division of Radiation Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, TWN.,Radiation Science and Proton Therapy Center, National Taiwan University College of Medicine, Taipei, TWN
| | - Yoshiko Oshiro
- Department of Radiation Oncology, Tsukuba Medical Center Hospital, Tsukuba, JPN
| | | | | | - Takashi Iizumi
- Department of Radiation Oncology, Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba, JPN
| | - Haruko Numajiri
- Department of Radiation Oncology, Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba, JPN
| | - Kei Nakai
- Department of Radiation Oncology, University of Tsukuba Hospital, Tsukuba, JPN
| | - Toshiyuki Okumura
- Department of Radiation Oncology, Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba, JPN
| | | | | |
Collapse
|
57
|
Kasper J, Frydrychowicz C, Jähne K, Wende T, Wilhelmy F, Arlt F, Seidel C, Hoffmann KT, Meixensberger J. The Role of Delayed Radiotherapy Initiation in Patients with Newly Diagnosed Glioblastoma with Residual Tumor Mass. J Neurol Surg A Cent Eur Neurosurg 2021; 83:252-258. [PMID: 34496417 DOI: 10.1055/s-0041-1730965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Treatment for newly diagnosed isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM) includes maximum safe resection, followed by adjuvant radio(chemo)therapy (RCx) with temozolomide. There is evidence that it is safe for GBM patients to prolong time to irradiation over 4 weeks after surgery. This study aimed at evaluating whether this applies to GBM patients with different levels of residual tumor volume (RV). METHODS Medical records of all patients with newly diagnosed GBM at our department between 2014 and 2018 were reviewed. Patients who received adjuvant radio (chemo) therapy, aged older than 18 years, and with adequate perioperative imaging were included. Initial and residual tumor volumes were determined. Time to irradiation was dichotomized into two groups (≤28 and >28 days). Univariate analysis with Kaplan-Meier estimate and log-rank test was performed. Survival prediction and multivariate analysis were performed employing Cox proportional hazard regression. RESULTS One hundred and twelve patients were included. Adjuvant treatment regimen, extent of resection, residual tumor volume, and O6-methylguanine DNA methyltransferase (MGMT) promoter methylation were statistically significant factors for overall survival (OS). Time to irradiation had no impact on progression-free survival (p = 0.946) or OS (p = 0.757). When stratified for different thresholds of residual tumor volume, survival predication via Cox regression favored time to irradiation below 28 days for patients with residual tumor volume above 2 mL, but statistical significance was not reached. CONCLUSION Time to irradiation had no significant influence on OS of the entire cohort. Nevertheless, a statistically nonsignificant survival prolongation could be observed in patients with residual tumor volume > 2 mL when admitted to radiotherapy within 28 days after surgery.
Collapse
Affiliation(s)
- Johannes Kasper
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Germany
| | - Clara Frydrychowicz
- Department of Neuropathology, University Hospital Leipzig, Leipzig, Sachsen, Germany
| | - Katja Jähne
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Germany
| | - Tim Wende
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Germany
| | - Florian Wilhelmy
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Germany
| | - Felix Arlt
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Germany
| | - Clemens Seidel
- Department of Radio-Oncology, University Hospital Leipzig, Leipzig, Sachsen, Germany
| | - Karl-Titus Hoffmann
- Department of Neuro-Radiology, University Hospital Leipzig, Leipzig, Sachsen, Germany
| | | |
Collapse
|
58
|
Ruiz-Garcia H, Ramirez-Loera C, Malouff TD, Seneviratne DS, Palmer JD, Trifiletti DM. Novel Strategies for Nanoparticle-Based Radiosensitization in Glioblastoma. Int J Mol Sci 2021; 22:9673. [PMID: 34575840 PMCID: PMC8465220 DOI: 10.3390/ijms22189673] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/09/2023] Open
Abstract
Radiotherapy (RT) is one of the cornerstones in the current treatment paradigm for glioblastoma (GBM). However, little has changed in the management of GBM since the establishment of the current protocol in 2005, and the prognosis remains grim. Radioresistance is one of the hallmarks for treatment failure, and different therapeutic strategies are aimed at overcoming it. Among these strategies, nanomedicine has advantages over conventional tumor therapeutics, including improvements in drug delivery and enhanced antitumor properties. Radiosensitizing strategies using nanoparticles (NP) are actively under study and hold promise to improve the treatment response. We aim to describe the basis of nanomedicine for GBM treatment, current evidence in radiosensitization efforts using nanoparticles, and novel strategies, such as preoperative radiation, that could be synergized with nanoradiosensitizers.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| | | | - Timothy D. Malouff
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Danushka S. Seneviratne
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Joshua D. Palmer
- Department of Radiation Oncology, Ohio State University, Columbus, OH 43210, USA;
| | - Daniel M. Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
59
|
Liang HKT, Mizumoto M, Ishikawa E, Matsuda M, Tanaka K, Kohzuki H, Numajiri H, Oshiro Y, Okumura T, Matsumura A, Sakurai H. Peritumoral edema status of glioblastoma identifies patients reaching long-term disease control with specific progression patterns after tumor resection and high-dose proton boost. J Cancer Res Clin Oncol 2021; 147:3503-3516. [PMID: 34459971 PMCID: PMC8557163 DOI: 10.1007/s00432-021-03765-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/13/2021] [Indexed: 01/22/2023]
Abstract
Background Glioblastoma peritumoral edema (PE) extent is associated with survival and progression pattern after tumor resection and radiotherapy (RT). To increase tumor control, proton beam was adopted to give high-dose boost (> 90 Gy). However, the correlation between PE extent and prognosis of glioblastoma after postoperative high-dose proton boost (HDPB) therapy stays unknown. We intend to utilize the PE status to classify the survival and progression patterns. Methods Patients receiving HDPB (96.6 GyE) were retrospectively evaluated. Limited peritumoral edema (LPE) was defined as PE extent < 3 cm with a ratio of PE extent to tumor maximum diameter of < 0.75. Extended progressive disease (EPD) was defined as progression of tumors extending > 1 cm from the tumor bed edge. Results After long-term follow-up (median 88.7, range 63.6–113.8 months) for surviving patients with (n = 13) and without (n = 32) LPE, the median overall survival (OS) and progression-free survival (PFS) were 77.2 vs. 16.7 months (p = 0.004) and 13.6 vs. 8.6 months (p = 0.02), respectively. In multivariate analyses combined with factors of performance, age, tumor maximum diameter, and tumor resection extent, LPE remained a significant factor for favorable OS and PFS. The rates of 5-year complete response, EPD, and distant metastasis with and without LPE were 38.5% vs. 3.2% (p = 0.005), 7.7% vs. 40.6% (p = 0.04), and 0% vs. 34.4% (p = 0.02), respectively. Conclusions The LPE status effectively identified patients with relative long-term control and specific progression patterns after postoperative HDPB for glioblastoma. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03765-6.
Collapse
Affiliation(s)
- Hsiang-Kuang Tony Liang
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Department of Radiation Oncology, National Taiwan University Cancer Center, National Taiwan University Hospital, Taipei, Taiwan
- Division of Radiation Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Masashi Mizumoto
- Department of Radiation Oncology, Proton Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Eiichi Ishikawa
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masahide Matsuda
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Keiichi Tanaka
- Department of Radiation Oncology, Proton Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hidehiro Kohzuki
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruko Numajiri
- Department of Radiation Oncology, Proton Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiko Oshiro
- Department of Radiation Oncology, Tsukuba Medical Center Hospital, Tsukuba, Ibaraki, Japan
| | - Toshiyuki Okumura
- Department of Radiation Oncology, Proton Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Matsumura
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hideyuki Sakurai
- Department of Radiation Oncology, Proton Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
60
|
Laack NN, Pafundi D, Anderson SK, Kaufmann T, Lowe V, Hunt C, Vogen D, Yan E, Sarkaria J, Brown P, Kizilbash S, Uhm J, Ruff M, Zakhary M, Zhang Y, Seaberg M, Wan Chan Tseung HS, Kabat B, Kemp B, Brinkmann D. Initial Results of a Phase 2 Trial of 18F-DOPA PET-Guided Dose-Escalated Radiation Therapy for Glioblastoma. Int J Radiat Oncol Biol Phys 2021; 110:1383-1395. [PMID: 33771703 DOI: 10.1016/j.ijrobp.2021.03.032] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/21/2021] [Accepted: 03/16/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Our previous work demonstrated that 3,4-dihydroxy-6-[18F]-fluoro-L-phenylalanine (18F-DOPA) positron emission tomography (PET) is sensitive and specific for identifying regions of high density and biologically aggressive glioblastoma. The purpose of this prospective phase 2 study was to determine the safety and efficacy of biologic-guided, dose-escalated radiation therapy (DERT) using 18F-DOPA PET in patients with glioblastoma. METHODS AND MATERIALS Patients with newly diagnosed, histologically confirmed glioblastoma aged ≥18 years without contraindications to 18F-DOPA were eligible. Target volumes included 51, 60, and 76 Gy in 30 fractions with a simultaneous integrated boost, and concurrent and adjuvant temozolomide for 6 months. 18F-DOPA PET imaging was used to guide DERT. The study was designed to detect a true progression-free survival (PFS) at 6 months (PFS6) rate ≥72.5% in O6-methylguanine methyltransferase (MGMT) unmethylated patients (DE-Un), with an overall significance level (alpha) of 0.20 and a power of 80%. Kaplan-Meier analysis was performed for PFS and overall survival (OS). Historical controls (HCs) included 139 patients (82 unmethylated) treated on prospective clinical trials or with standard RT at our institution. Toxicities were evaluated with Common Terminology Criteria for Adverse Events v4.0. RESULTS Between January 2014 and December 2018, 75 evaluable patients were enrolled (39 DE-Un, 24 methylated [DE-Mth], and 12 indeterminate). PFS6 for DE-Un was 79.5% (95% confidence interval, 63.1%-90.1%). Median PFS was longer for DE-Un patients compared with historical controls (8.7 months vs 6.6 months; P = .017). OS was similarly longer, but the difference was not significant (16.0 vs 13.5 months; P = .13). OS was significantly improved for DE-Mth patients compared with HC-Mth (35.5 vs 23.3 months; P = .049) despite nonsignificant improvement in PFS (10.7 vs 9.0 months; P = .26). Grade 3 central nervous system necrosis occurred in 13% of patients, but treatment with bevacizumab improved symptoms in all cases. CONCLUSIONS 18F-DOPA PET-guided DERT appears to be safe, and it significantly improves PFS in MGMT unmethylated glioblastoma. OS is significantly improved in MGMT methylated patients. Further investigation of 18F-DOPA PET biologic guided DERT for glioblastoma is warranted.
Collapse
Affiliation(s)
| | - Deanna Pafundi
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida
| | | | | | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | - Diane Vogen
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Elizabeth Yan
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jann Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Paul Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Sani Kizilbash
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Joon Uhm
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Michael Ruff
- Deptartment of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Mark Zakhary
- Department of Radiation Oncology, University of Maryland, Baltimore, Maryland
| | - Yan Zhang
- Department of Research, Mayo Clinic, Jacksonville, Florida
| | | | | | - Brian Kabat
- Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Bradley Kemp
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Debra Brinkmann
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
61
|
Adegboyega G, Kanmounye US, Petrinic T, Ozair A, Bandyopadhyay S, Kuri A, Zolo Y, Marks K, Ramjee S, Baticulon RE, Vaqas B, on behalf of InciSion UK Collaborative. Global Landscape of Glioblastoma Multiforme Management in the Stupp Protocol Era: Systematic Review Protocol. Int J Surg Protoc 2021; 25:108-113. [PMID: 34222733 PMCID: PMC8231457 DOI: 10.29337/ijsp.148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/06/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme is the most common and aggressive primary adult brain neoplasm. The current standard of care is maximal safe surgical resection, radiotherapy with concomitant temozolomide, followed by adjuvant temozolomide according to the Stupp protocol. Although the protocol is well adopted in high-income countries (HICs), little is known about its adoption in low- and middle-income countries (LMICs). The aim of this study is to describe a protocol design for a systematic review of published studies outlining the differences in GBM management between HICs and LMICs. METHODS A systematic review will be conducted. MedLine via Ovid, Embase and Global Index Medicus will be searched from inception to date in order to identify the relevant studies. Adult patients (>18 years) with histologically confirmed primary unifocal GBM will be included. Surgical and chemoradiation management of GBM tumours will be considered. Commentaries, original research, non-peer reviewed pieces, opinion pieces, editorials and case reports will be included. RESULTS Primary outcomes will include rates of complications, disability-adjusted life years (DALYs), prognosis, progression-free survival (PFS), overall survival (OS) as well as rate of care abandonment and delay. Secondary outcomes will include the presence of neuro-oncology subspecialty training programs. DISCUSSION This systematic review will be the first to compare the current landscape of GBM management in HICs and LMICs, highlighting pertinent themes that may be used to optimise treatment in both financial brackets. SYSTEMATIC REVIEW REGISTRATION The protocol has been registered on the International Prospective Register of Systematic Reviews (PROSPERO; registration number: CRD42020215843). HIGHLIGHTS Glioblastoma multiforme (GBM) remains the most common primary adult cerebral neoplasm, with an age-adjusted incidence rate of 3.22 per 100,000 population and a 5-year survival rate of 6.8%Despite the well-evidenced efficacy of Stupp protocol, the implementation of this approach bears an institutional and individual financial burden that is particularly notable in low- and middle-income countries (LMICs)This systematic review will be the first to compare the current landscape of GBM management in HICs and LMICs, highlighting pertinent themes that may be used to optimise treatment in both financial brackets.
Collapse
Affiliation(s)
- Gideon Adegboyega
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, United Kingdom
- Research Department, Association of Future African Neurosurgeons, Yaounde, Cameroon
| | | | - Tatjana Petrinic
- Bodleian Health Care Libraries, University of Oxford, Oxfordshire, United Kingdom
| | - Ahmad Ozair
- Faculty of Medicine, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Soham Bandyopadhyay
- Oxford University Global Surgery Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxfordshire, United Kingdom
| | - Ashvin Kuri
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Yvan Zolo
- Research Department, Association of Future African Neurosurgeons, Yaounde, Cameroon
- Faculty of Health Sciences, University of Buea, Buea, Cameroon
| | - Katya Marks
- Medical Sciences Division, John Radcliffe Hospital, University of Oxford, OX3 9DU Oxfordshire, Oxford, United Kingdom
| | - Serena Ramjee
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Ronnie E. Baticulon
- Division of Neurosurgery, Department of Neurosciences, Philippine General Hospital, Manila, Philippines
- Department of Anatomy, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Babar Vaqas
- Department of Neurosurgery, Queens Hospital, Romford, United Kingdom
- Imperial College London, London, United Kingdom
| | | |
Collapse
|
62
|
Biamonte F, Sica G, Filippini A, D’Alessio A. Evidence of Reelin Signaling in GBM and Its Derived Cancer Stem Cells. Brain Sci 2021; 11:brainsci11060745. [PMID: 34205192 PMCID: PMC8227261 DOI: 10.3390/brainsci11060745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 11/22/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive and malignant form of primary brain cancer, characterized by an overall survival time ranging from 12 to 18 months. Despite the progress in the clinical treatment and the growing number of experimental data aimed at investigating the molecular bases of GBM development, the disease remains characterized by a poor prognosis. Recent studies have proposed the existence of a population of GBM cancer stem cells (CSCs) endowed with self-renewal capability and a high tumorigenic potential that are believed to be responsible for the resistance against common chemotherapy and radiotherapy treatments. Reelin is a large secreted extracellular matrix glycoprotein, which contributes to positioning, migration, and laminar organization of several central nervous system structures during brain development. Mutations of the reelin gene have been linked to disorganization of brain structures during development and behavioral anomalies. In this study, we explored the expression of reelin in GBM and its related peritumoral tissue and performed the same analysis in CSCs isolated from both GBM (GCSCs) and peritumoral tissue (PCSCs) of human patients. Our findings reveal (i) the higher expression of reelin in GBM compared to the peritumoral tissue by immunohistochemical analysis, (ii) the mRNA expression of both reelin and its adaptor molecule Dab1 in either CSC subtypes, although at a different extent; and (iii) the contribution of CSCs-derived reelin in the migration of human primary GBM cell line U87MG. Taken together, our data indicate that the expression of reelin in GBM may represent a potential contribution to the regulation of GBM cancer stem cells behavior, further stimulating the interest on the reelin pathway as a potential target for GBM treatment.
Collapse
Affiliation(s)
- Filippo Biamonte
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italy;
| | - Gigliola Sica
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italy;
| | - Antonio Filippini
- Dipartimento di Scienze Anatomiche, Istologiche, Medico Legali e dell’Apparato Locomotore, Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy;
| | - Alessio D’Alessio
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italy;
- Correspondence:
| |
Collapse
|
63
|
Adjuvant Temozolomide for the Treatment of Glioblastoma: A Meta-analysis of Randomized Controlled Studies. Clin Neuropharmacol 2021; 44:132-137. [PMID: 34039842 DOI: 10.1097/wnf.0000000000000458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION The efficacy of adjuvant temozolomide to radiotherapy for glioblastoma remained elusive. This meta-analysis aimed to explore the influence of radiotherapy plus adjuvant temozolomide on the efficacy and safety for glioblastoma. METHODS We have searched several databases including PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases through November 2020 and included randomized controlled trials assessing the efficacy and safety of adjuvant temozolomide to radiotherapy for glioblastoma. RESULTS Seven randomized controlled trials and 1900 patients were included in the meta-analysis. Overall, compared with radiotherapy for glioblastoma, adjuvant temozolomide was associated with significantly increased survival rate [odds ratio (OR), 4.04; 95% confidence interval (CI), 2.61-6.24; P < 0.00001], median progression-free survival (mean difference, 0.55; 95% CI, 0.03-1.07; P = 0.04), and hematological complications (OR, 4.12; 95% CI, 1.43-11.88; P = 0.009), but revealed no remarkable influence on adverse events (OR, 0.87; 95% CI, 0.36-2.09; P = 0.75) or serious adverse events (OR, 2.20; 95% CI, 0.55-8.70; P = 0.26). CONCLUSIONS Adjuvant temozolomide in combination with radiotherapy may improve the treatment efficacy for glioblastoma.
Collapse
|
64
|
A New Era of Neuro-Oncology Research Pioneered by Multi-Omics Analysis and Machine Learning. Biomolecules 2021; 11:biom11040565. [PMID: 33921457 PMCID: PMC8070530 DOI: 10.3390/biom11040565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Although the incidence of central nervous system (CNS) cancers is not high, it significantly reduces a patient’s quality of life and results in high mortality rates. A low incidence also means a low number of cases, which in turn means a low amount of information. To compensate, researchers have tried to increase the amount of information available from a single test using high-throughput technologies. This approach, referred to as single-omics analysis, has only been partially successful as one type of data may not be able to appropriately describe all the characteristics of a tumor. It is presently unclear what type of data can describe a particular clinical situation. One way to solve this problem is to use multi-omics data. When using many types of data, a selected data type or a combination of them may effectively resolve a clinical question. Hence, we conducted a comprehensive survey of papers in the field of neuro-oncology that used multi-omics data for analysis and found that most of the papers utilized machine learning techniques. This fact shows that it is useful to utilize machine learning techniques in multi-omics analysis. In this review, we discuss the current status of multi-omics analysis in the field of neuro-oncology and the importance of using machine learning techniques.
Collapse
|
65
|
Evren AE, Yurttaş L, Ekselli B, Aksoy O, Akalin-Çiftçi G. Design and Efficient Synthesis of Novel 4,5-Dimethylthiazole-Hydrazone Derivatives and their Anticancer Activity. LETT DRUG DES DISCOV 2021. [DOI: 10.2174/1570180817999201022192937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background::
Recently, researchers have been warning about the increased mortality of
the various cancer types. Also, the lung adenocarcinoma and the glioma types are burning issues for
world's health due to late or wrong diagnosis and/or insufficient treatment methods. For this
purpose, our research group designed and synthesized novel 4,5-dimethyl thiazole-hydrazone
derivatives which were tested against cancer and normal cell lines to understand the structureactivity
relationship (SAR).
Method::
The lead compounds were obtained by reacting 2-(substituted aryl-2-ylmethylene)
hydrazin-1-carbothioamide with 3-chloro-2-butanone derivatives. The structural elucidation of the
compounds was performed by 1H-NMR, 13C-NMR, and LC/MS-IT-TOF spectral and elemental
analyses. The synthesized compounds were tested in vitro for the anticancer activity against A549
human lung adenocarcinoma and C6 rat glioma cells and investigated for which pathway to induce
cell death. Also, the docking study of the active compounds was achieved to understand the SAR.
Result and Discussion::
The targeted compounds (2a-2l) were synthesized successfully above 70% yields, and
the analysis findings proved their purity. In general, the results of activity studies displayed
significant effects against at least one cell line, except compounds 2e (indol-3-yl) and 2h
(4-dimethylaminophenyl). Furthermore, compounds 2b and 2f displayed potential anticancer
activity. With the help of molecular docking study, a potential selectivity of compound 2f was
observed for type II protein kinase. On the other hand, compound 2b interacted with the active site
nearly the same as Dasatinib. Therefore, these two compounds could be used as a base on
developing selective anticancer drugs.
Conclusion::
Pyridin-2-yl (2b) derivative was found to be a favorable molecule with high anticancer
potency against C6 and A549 cell lines. Additionally, 1-naphthyl (2f) derivative was a worthy
compound for potential selectivity. In future studies, it will be our priority to focus on developing
derivatives of these two compounds (2b and 2f) and elucidate their mechanisms.
Collapse
Affiliation(s)
- Asaf Evrim Evren
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir,Turkey
| | - Leyla Yurttaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir,,Turkey
| | - Büşra Ekselli
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir,Turkey
| | - Onur Aksoy
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir,Turkey
| | - Gülşen Akalin-Çiftçi
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir,Turkey
| |
Collapse
|
66
|
Yu Q, Xiao W, Sun S, Sohrabi A, Liang J, Seidlits SK. Extracellular Matrix Proteins Confer Cell Adhesion-Mediated Drug Resistance Through Integrin α v in Glioblastoma Cells. Front Cell Dev Biol 2021; 9:616580. [PMID: 33834020 PMCID: PMC8021872 DOI: 10.3389/fcell.2021.616580] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/26/2021] [Indexed: 12/25/2022] Open
Abstract
Chemotherapy resistance to glioblastoma (GBM) remains an obstacle that is difficult to overcome, leading to poor prognosis of GBM patients. Many previous studies have focused on resistance mechanisms intrinsic to cancer cells; the microenvironment surrounding tumor cells has been found more recently to have significant impacts on the response to chemotherapeutic agents. Extracellular matrix (ECM) proteins may confer cell adhesion-mediated drug resistance (CAMDR). Here, expression of the ECM proteins laminin, vitronectin, and fibronectin was assessed in clinical GBM tumors using immunohistochemistry. Then, patient-derived GBM cells grown in monolayers on precoated laminin, vitronectin, or fibronectin substrates were treated with cilengitide, an integrin inhibitor, and/or carmustine, an alkylating chemotherapy. Cell adhesion and viability were quantified. Transcription factor (TF) activities were assessed over time using a bioluminescent assay in which GBM cells were transduced with lentiviruses containing consensus binding sites for specific TFs linked to expression a firefly luciferase reporter. Apoptosis, mediated by p53, was analyzed by Western blotting and immunocytofluorescence. Integrin αv activation of the FAK/paxillin/AKT signaling pathway and effects on expression of the proliferative marker Ki67 were investigated. To assess effects of integrin αv activation of AKT and ERK pathways, which are typically deregulated in GBM, and expression of epidermal growth factor receptor (EGFR), which is amplified and/or mutated in many GBM tumors, shRNA knockdown was used. Laminin, vitronectin, and fibronectin were abundant in clinical GBM tumors and promoted CAMDR in GBM cells cultured on precoated substrates. Cilengitide treatment induced cell detachment, which was most pronounced for cells cultured on vitronectin. Cilengitide treatment increased cytotoxicity of carmustine, reversing CAMDR. ECM adhesion increased activity of NFκB and decreased that of p53, leading to suppression of p53-mediated apoptosis and upregulation of multidrug resistance gene 1 (MDR1; also known as ABCB1 or P-glycoprotein). Expression of Ki67 was correlative with activation of the integrin αv-mediated FAK/paxillin/AKT signaling pathway. EGFR expression increased with integrin αv knockdown GBM cells and may represent a compensatory survival mechanism. These results indicate that ECM proteins confer CAMDR through integrin αv in GBM cells.
Collapse
Affiliation(s)
- Qi Yu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weikun Xiao
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Songping Sun
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alireza Sohrabi
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jesse Liang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Stephanie K Seidlits
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States.,Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, United States.,Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
67
|
Fisher JP, Adamson DC. Current FDA-Approved Therapies for High-Grade Malignant Gliomas. Biomedicines 2021; 9:biomedicines9030324. [PMID: 33810154 PMCID: PMC8004675 DOI: 10.3390/biomedicines9030324] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/26/2022] Open
Abstract
The standard of care (SOC) for high-grade gliomas (HGG) is maximally safe surgical resection, followed by concurrent radiation therapy (RT) and temozolomide (TMZ) for 6 weeks, then adjuvant TMZ for 6 months. Before this SOC was established, glioblastoma (GBM) patients typically lived for less than one year after diagnosis, and no adjuvant chemotherapy had demonstrated significant survival benefits compared with radiation alone. In 2005, the Stupp et al. randomized controlled trial (RCT) on newly diagnosed GBM patients concluded that RT plus TMZ compared to RT alone significantly improved overall survival (OS) (14.6 vs. 12.1 months) and progression-free survival (PFS) at 6 months (PFS6) (53.9% vs. 36.4%). Outside of TMZ, there are four drugs and one device FDA-approved for the treatment of HGGs: lomustine, intravenous carmustine, carmustine wafer implants, bevacizumab (BVZ), and tumor treatment fields (TTFields). These treatments are now mainly used to treat recurrent HGGs and symptoms. TTFields is the only treatment that has been shown to improve OS (20.5 vs. 15.6 months) and PFS6 (56% vs. 37%) in comparison to the current SOC. TTFields is the newest addition to this list of FDA-approved treatments, but has not been universally accepted yet as part of SOC.
Collapse
Affiliation(s)
- Jacob P. Fisher
- Division of Biochemistry, Southern Virginia University, Buena Vista, VA 24416, USA
- Correspondence:
| | - David C. Adamson
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA;
- Atlanta VA Healthcare System, Decatur, GA 30033, USA
| |
Collapse
|
68
|
Hasbum A, Quintanilla J, Jr JA, Ding MH, Levy A, Chew SA. Strategies to better treat glioblastoma: antiangiogenic agents and endothelial cell targeting agents. Future Med Chem 2021; 13:393-418. [PMID: 33399488 PMCID: PMC7888526 DOI: 10.4155/fmc-2020-0289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive form of glioma, with poor prognosis and high mortality rates. As GBM is a highly vascularized cancer, antiangiogenic therapies to halt or minimize the rate of tumor growth are critical to improving treatment. In this review, antiangiogenic therapies, including small-molecule drugs, nucleic acids and proteins and peptides, are discussed. The authors further explore biomaterials that have been utilized to increase the bioavailability and bioactivity of antiangiogenic factors for better antitumor responses in GBM. Finally, the authors summarize the current status of biomaterial-based targeting moieties that target endothelial cells in GBM to more efficiently deliver therapeutics to these cells and avoid off-target cell or organ side effects.
Collapse
Affiliation(s)
- Asbiel Hasbum
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA
| | - Jaqueline Quintanilla
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - Juan A Amieva Jr
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - May-Hui Ding
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - Arkene Levy
- Dr Kiran C Patel College of Allopathic Medicine, Nova Southeastern University, FL 33314, USA
| | - Sue Anne Chew
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| |
Collapse
|
69
|
Randomized Controlled Immunotherapy Clinical Trials for GBM Challenged. Cancers (Basel) 2020; 13:cancers13010032. [PMID: 33374196 PMCID: PMC7796083 DOI: 10.3390/cancers13010032] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Although multiple meta-analyses on active specific immunotherapy treatment for glioblastoma multiforme (GBM) have demonstrated a significant prolongation of overall survival, no single research group has succeeded in demonstrating the efficacy of this type of treatment in a prospective, double-blind, placebo-controlled, randomized clinical trial. In this paper, we explain how the complexity of the tumor biology and tumor–host interactions make proper stratification of a control group impossible. The individualized characteristics of advanced therapy medicinal products for immunotherapy contribute to heterogeneity within an experimental group. The dynamics of each tumor and in each patient aggravate comparative stable patient groups. Finally, combinations of immunotherapy strategies should be integrated with first-line treatment. We illustrate the complexity of a combined first-line treatment with individualized multimodal immunotherapy in a group of 70 adults with GBM and demonstrate that the integration of immunogenic cell death treatment within maintenance chemotherapy followed by dendritic cell vaccines and maintenance immunotherapy might provide a step towards improving the overall survival rate of GBM patients. Abstract Immunotherapies represent a promising strategy for glioblastoma multiforme (GBM) treatment. Different immunotherapies include the use of checkpoint inhibitors, adoptive cell therapies such as chimeric antigen receptor (CAR) T cells, and vaccines such as dendritic cell vaccines. Antibodies have also been used as toxin or radioactive particle delivery vehicles to eliminate target cells in the treatment of GBM. Oncolytic viral therapy and other immunogenic cell death-inducing treatments bridge the antitumor strategy with immunization and installation of immune control over the disease. These strategies should be included in the standard treatment protocol for GBM. Some immunotherapies are individualized in terms of the medicinal product, the immune target, and the immune tumor–host contact. Current individualized immunotherapy strategies focus on combinations of approaches. Standardization appears to be impossible in the face of complex controlled trial designs. To define appropriate control groups, stratification according to the Recursive Partitioning Analysis classification, MGMT promotor methylation, epigenetic GBM sub-typing, tumor microenvironment, systemic immune functioning before and after radiochemotherapy, and the need for/type of symptom-relieving drugs is required. Moreover, maintenance of a fixed treatment protocol for a dynamic, deadly cancer disease in a permanently changing tumor–host immune context might be inappropriate. This complexity is illustrated using our own data on individualized multimodal immunotherapies for GBM. Individualized medicines, including multimodal immunotherapies, are a rational and optimal yet also flexible approach to induce long-term tumor control. However, innovative methods are needed to assess the efficacy of complex individualized treatments and implement them more quickly into the general health system.
Collapse
|
70
|
Valiyaveettil D, Malik M, Joseph D, Ahmed SF, Kothwal SA. Prognostic factors and outcomes in anaplastic gliomas: An institutional experience. South Asian J Cancer 2020; 7:1-4. [PMID: 29600221 PMCID: PMC5865085 DOI: 10.4103/sajc.sajc_55_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background: There is lack of clear evidence and treatment guidelines for anaplastic gliomas (AGs) with very few studies focusing exclusively on these patients. The aim of the study was to analyze the clinical profile and survival in these patients. Materials and Methods: Patients of AGs treated with radiation and concurrent ± adjuvant chemotherapy from January 2010 to December 2015 were analyzed. Statistical analysis was done using SPSS version 20 software. Results: A total of 100 patients were included in the study. The median age was 35 years (range 6–68 years). Eighty-four patients had follow-up details and were included for survival analysis. The 5-year overall survival (OS) was 58%. Age, presentation with seizures, and focal neurological deficit were not found to significantly influence survival. The 5-year survival for oligodendroglioma and astrocytoma was 69% and 52%, respectively. Patients with Karnofsky Performance Score (KPS) of ≥70 had a significantly better 5-year OS (65%) as compared to those with KPS <70 (33%) (P = 0.000). The use of adjuvant temozolomide (TMZ) showed longer 5-year OS of 67.7% compared to 36% in patients who did not receive adjuvant chemotherapy (P = 0.018). Patients receiving both concurrent and adjuvant TMZ showed longer 5-year OS (68.5% vs. 40%, P = 0.010). Twenty-two patients had recurrence with average time to recurrence being 37 months. Fourteen patients underwent salvage surgery and two patients received reirradiation. Conclusions: OS significantly correlated with KPS and receipt of concurrent and adjuvant chemotherapy with TMZ. Therefore, adjuvant radiation with concurrent and adjuvant TMZ should be the standard of care for AGs.
Collapse
Affiliation(s)
- Deepthi Valiyaveettil
- Department of Radiation Oncology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Monica Malik
- Department of Radiation Oncology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Deepa Joseph
- Department of Radiation Oncology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Syed Fayaz Ahmed
- Department of Radiation Oncology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Syed Akram Kothwal
- Department of Radiation Oncology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| |
Collapse
|
71
|
EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol 2020; 18:170-186. [PMID: 33293629 PMCID: PMC7904519 DOI: 10.1038/s41571-020-00447-z] [Citation(s) in RCA: 1045] [Impact Index Per Article: 209.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 01/16/2023]
Abstract
In response to major changes in diagnostic algorithms and the publication of mature results from various large clinical trials, the European Association of Neuro-Oncology (EANO) recognized the need to provide updated guidelines for the diagnosis and management of adult patients with diffuse gliomas. Through these evidence-based guidelines, a task force of EANO provides recommendations for the diagnosis, treatment and follow-up of adult patients with diffuse gliomas. The diagnostic component is based on the 2016 update of the WHO Classification of Tumors of the Central Nervous System and the subsequent recommendations of the Consortium to Inform Molecular and Practical Approaches to CNS Tumour Taxonomy — Not Officially WHO (cIMPACT-NOW). With regard to therapy, we formulated recommendations based on the results from the latest practice-changing clinical trials and also provide guidance for neuropathological and neuroradiological assessment. In these guidelines, we define the role of the major treatment modalities of surgery, radiotherapy and systemic pharmacotherapy, covering current advances and cognizant that unnecessary interventions and expenses should be avoided. This document is intended to be a source of reference for professionals involved in the management of adult patients with diffuse gliomas, for patients and caregivers, and for health-care providers. Herein, the European Association of Neuro-Oncology (EANO) provides recommendations for the diagnosis, treatment and follow-up of adult patients with diffuse gliomas. These evidence-based guidelines incorporate major changes in diagnostic algorithms based on the 2016 update of the WHO Classification of Tumors of the Central Nervous System as well as on evidence from recent large clinical trials.
Collapse
|
72
|
Nabors LB, Portnow J, Ahluwalia M, Baehring J, Brem H, Brem S, Butowski N, Campian JL, Clark SW, Fabiano AJ, Forsyth P, Hattangadi-Gluth J, Holdhoff M, Horbinski C, Junck L, Kaley T, Kumthekar P, Loeffler JS, Mrugala MM, Nagpal S, Pandey M, Parney I, Peters K, Puduvalli VK, Robins I, Rockhill J, Rusthoven C, Shonka N, Shrieve DC, Swinnen LJ, Weiss S, Wen PY, Willmarth NE, Bergman MA, Darlow SD. Central Nervous System Cancers, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 18:1537-1570. [PMID: 33152694 DOI: 10.6004/jnccn.2020.0052] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Guidelines for Central Nervous System (CNS) Cancers focus on management of adult CNS cancers ranging from noninvasive and surgically curable pilocytic astrocytomas to metastatic brain disease. The involvement of an interdisciplinary team, including neurosurgeons, radiation therapists, oncologists, neurologists, and neuroradiologists, is a key factor in the appropriate management of CNS cancers. Integrated histopathologic and molecular characterization of brain tumors such as gliomas should be standard practice. This article describes NCCN Guidelines recommendations for WHO grade I, II, III, and IV gliomas. Treatment of brain metastases, the most common intracranial tumors in adults, is also described.
Collapse
Affiliation(s)
| | | | - Manmeet Ahluwalia
- 3Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Henry Brem
- 5The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Steven Brem
- 6Abramson Cancer Center at the University of Pennsylvania
| | | | - Jian L Campian
- 8Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | | | - Craig Horbinski
- 13Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Larry Junck
- 14University of Michigan Rogel Cancer Center
| | | | - Priya Kumthekar
- 13Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | - Manjari Pandey
- 19St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | | | | | - Vinay K Puduvalli
- 21The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Ian Robins
- 22University of Wisconsin Carbone Cancer Center
| | - Jason Rockhill
- 23Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | | | - Lode J Swinnen
- 5The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | | |
Collapse
|
73
|
Profyris C, Chen E, Young IM, Chendeb K, Ahsan SA, Briggs RG, Sughrue ME, Teo C. Anaplastic Oligodendroglioma - Is Adjuvant Radiotherapy Mandatory following Maximal Surgical Resection?: Grade 3 Oligo Radiotherapy. Clin Neurol Neurosurg 2020; 200:106303. [PMID: 33109468 DOI: 10.1016/j.clineuro.2020.106303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/25/2020] [Accepted: 10/11/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Current anaplastic oligodendroglioma (AO) management strategies involve surgical resection followed by adjuvant radiotherapy and/or chemotherapy. We investigated a subset of patients at our institution with AO, who, based on their treatment preferences, received surgery without any form of adjuvant therapy. This subset of patients was compared to a cohort with AO who received adjuvant therapy in order to investigate any differences in clinical and survival outcomes. METHODS A retrospective review of all AO patients treated by the senior author was undertaken between 1994 and 2018. A total of thirty-three cases were identified. Eleven had surgery alone, and twenty-two had surgery with adjuvant therapy. Progression free (PFS) and overall survival (OS) were compared between cohorts and potential confounders were addressed. RESULTS Gross total resection was achieved in 29 patients, and near total resection in 4 patients. PFS was not statistically different between patients treated with surgery alone versus patients receiving surgery plus adjuvant therapy (surgery alone: 84 ± 16 months; surgery with radiotherapy: 60 ± 9 months; p = 0.08). In addition, OS was also not statistically different between these groups (surgery alone: 215 ± 17 months; surgery with therapy: 241 ± 22 months; p = 0.44). CONCLUSIONS It is reasonable to consider a "watch and monitor" surveillance strategy in patients who decline adjuvant radiotherapy following surgical resection of their AO. Patients should be made aware that this treatment plan is not standard within current models of care for AO.
Collapse
Affiliation(s)
- Christos Profyris
- Department of Neurosurgery, Prince of Wales Private Hospital, Randwick, NSW, Australia; Department of Neurosurgery, Helen Joseph Hospital, University of the Witwatersrand, Auckland Park, Johannesburg, South Africa
| | - Emily Chen
- Department of Neurosurgery, Prince of Wales Private Hospital, Randwick, NSW, Australia
| | | | - Kassem Chendeb
- Department of Neurosurgery, Prince of Wales Private Hospital, Randwick, NSW, Australia
| | - Syed A Ahsan
- Department of Neurosurgery, Prince of Wales Private Hospital, Randwick, NSW, Australia
| | - Robert G Briggs
- Department of Neurosurgery, University of Southern California, Los Angeles, California
| | - Michael E Sughrue
- Department of Neurosurgery, Prince of Wales Private Hospital, Randwick, NSW, Australia.
| | - Charles Teo
- Department of Neurosurgery, Prince of Wales Private Hospital, Randwick, NSW, Australia
| |
Collapse
|
74
|
Choi J, Kim G, Cho SB, Im HJ. Radiosensitizing high-Z metal nanoparticles for enhanced radiotherapy of glioblastoma multiforme. J Nanobiotechnology 2020; 18:122. [PMID: 32883290 PMCID: PMC7470617 DOI: 10.1186/s12951-020-00684-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an essential step during the treatment of glioblastoma multiforme (GBM), one of the most lethal malignancies. The survival in patients with GBM was improved by the current standard of care for GBM established in 2005 but has stagnated since then. Since GBM is a radioresistant malignancy and the most of GBM recurrences occur in the radiotherapy field, increasing the effectiveness of radiotherapy using high-Z metal nanoparticles (NPs) has recently attracted attention. This review summarizes the progress in radiotherapy approaches for the current treatment of GBM, the physical and biological mechanisms of radiosensitization through high-Z metal NPs, and the results of studies on radiosensitization in the in vitro and in vivo GBM models using high-Z metal NPs to date.
Collapse
Affiliation(s)
- Jinyeong Choi
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Gaeun Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Su Bin Cho
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hyung-Jun Im
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
75
|
Caccese M, Padovan M, D'Avella D, Chioffi F, Gardiman MP, Berti F, Busato F, Bellu L, Bergo E, Zoccarato M, Fassan M, Zagonel V, Lombardi G. Anaplastic Astrocytoma: State of the art and future directions. Crit Rev Oncol Hematol 2020; 153:103062. [PMID: 32717623 DOI: 10.1016/j.critrevonc.2020.103062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/17/2020] [Accepted: 07/12/2020] [Indexed: 01/05/2023] Open
Abstract
Anaplastic Astrocytoma(AA) is a malignant, diffusely infiltrating, primary brain tumor. According to the WHO 2016 classification of central-nervous-system tumors, AA has been described as a glial tumor with no co-deletion of 1p/19q, and is divided into IDH mutated tumor, characterized by better prognosis, and IDH wild-type form, with worse prognosis. The standard of care is maximal safe resection followed by radiotherapy and chemotherapy with temozolomide. Several efforts have been made to evaluate, according to molecular selection, which is the best post-surgical treatment. At recurrence, the treatment remains challenging and some trials are ongoing to evaluate new potential drugs, alone or in combination with chemotherapy. We performed a description of the status of the art on diagnosis, molecular characteristics and treatment of AA. In particular, we focused our details on new drugs; indeed, a deeper knowledge of the molecular characteristics of gliomas could lead to to development of active personalized treatments according with precision medicine.
Collapse
Affiliation(s)
- Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy; Clinical and Experimental Oncology and Immunology PhD Program, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.
| | - Marta Padovan
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Domenico D'Avella
- Accademic Neurosurgery, Department of Neurosciences, University of Padua Medical School, Padua, Italy
| | - Franco Chioffi
- Department of Neurosurgery, Padua University Hospital, Padua, Italy
| | - Marina Paola Gardiman
- Surgical Pathology Unit, Department of Medicine (DIMED), University Hospital of Padua, Padua, Italy
| | - Franco Berti
- Radiation Therapy and Nuclear Medicine Unit, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Fabio Busato
- Radiation Therapy and Nuclear Medicine Unit, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Luisa Bellu
- Radiation Therapy and Nuclear Medicine Unit, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Eleonora Bergo
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Marco Zoccarato
- Department of Neurology, Ospedale S. Antonio, Azienda Ospedaliera Di Padova, Padua, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University Hospital of Padua, Padua, Italy
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology - IRCCS, Padua, Italy
| |
Collapse
|
76
|
Biotherapy of Brain Tumors with Phosphatidylserine-Targeted Radioiodinated SapC-DOPS Nanovesicles. Cells 2020; 9:cells9091960. [PMID: 32854321 PMCID: PMC7565346 DOI: 10.3390/cells9091960] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme (GBM), a common type of brain cancer, has a very poor prognosis. In general, viable GBM cells exhibit elevated phosphatidylserine (PS) on their membrane surface compared to healthy cells. We have developed a drug, saposin C-dioleoylphosphatidylserine (SapC-DOPS), that selectively targets cancer cells by honing in on this surface PS. To examine whether SapC-DOPS, a stable, blood–brain barrier-penetrable nanovesicle, could be an effective delivery system for precise targeted therapy of radiation, we iodinated several carbocyanine-based fluorescent reporters with either stable iodine (127I) or radioactive isotopes (125I and 131I). While all of the compounds, when incorporated into the SapC-DOPS delivery system, were taken up by human GBM cell lines, we chose the two that best accumulated in the cells (DiI (22,3) and DiD (16,16)). Pharmacokinetics were conducted with 125I-labeled compounds and indicated that DiI (22,3)-SapC-DOPS had a time to peak in the blood of 0.66 h and an elimination half-life of 8.4 h. These values were 4 h and 11.5 h, respectively, for DiD (16,16)-SapC-DOPS. Adult nude mice with GBM cells implanted in their brains were treated with 131I-DID (16,16)-SapC-DOPS. Mice receiving the radionuclide survived nearly 50% longer than the control groups. These data suggest a potential novel, personalized treatment for a devastating brain disease.
Collapse
|
77
|
Vargas López AJ, Fernández Carballal C, Valera Melé M, Rodríguez-Boto G. Survival analysis in high-grade glioma: the role of salvage surgery. Neurologia 2020; 38:S0213-4853(20)30125-0. [PMID: 32709508 DOI: 10.1016/j.nrl.2020.04.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 03/21/2020] [Accepted: 04/01/2020] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVES This study addresses the survival of consecutive patients with high-grade gliomas treated at the same institution over a period of 10 years. We analyse the importance of associated factors and the role of salvage surgery at the time of progression. METHODS We retrospectively analysed a series of patients with World Health Organization (WHO) grade III/IV gliomas treated between 2008 and 2017 at Hospital Gregorio Marañón (Madrid, Spain). Clinical, radiological, and anatomical pathology data were obtained from patient clinical histories. RESULTS Follow-up was completed in 233 patients with HGG. Mean age was 62.2 years. The median survival time was 15.4 months. Of 133 patients (59.6%) who had undergone surgery at the time of diagnosis, 43 (32.3%) underwent salvage surgery at the time of progression. This subgroup presented longer overall survival and survival after progression. Higher Karnofsky Performance Status score at diagnosis, a greater extent of surgical resection, and initial diagnosis of WHO grade III glioma were also associated with longer survival. CONCLUSIONS About one-third of patients with HGG may be eligible for salvage surgery at the time of progression. Salvage surgery in this subgroup of patients was significantly associated with longer survival.
Collapse
Affiliation(s)
- A J Vargas López
- Servicio de Neurocirugía, Hospital Universitario Torrecárdenas, Almería, España; Programa de Doctorado en Medicina y Cirugía, Universidad Autónoma de Madrid, Madrid, España.
| | - C Fernández Carballal
- Servicio de Neurocirugía, Hospital General Universitario Gregorio Marañón, Madrid, España
| | - M Valera Melé
- Servicio de Neurocirugía, Hospital General Universitario Gregorio Marañón, Madrid, España
| | - G Rodríguez-Boto
- Programa de Doctorado en Medicina y Cirugía, Universidad Autónoma de Madrid, Madrid, España; Servicio de Neurocirugía, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, España
| |
Collapse
|
78
|
Khoshnevis M, Carozzo C, Brown R, Bardiès M, Bonnefont-Rebeix C, Belluco S, Nennig C, Marcon L, Tillement O, Gehan H, Louis C, Zahi I, Buronfosse T, Roger T, Ponce F. Feasibility of intratumoral 165Holmium siloxane delivery to induced U87 glioblastoma in a large animal model, the Yucatan minipig. PLoS One 2020; 15:e0234772. [PMID: 32555746 PMCID: PMC7302492 DOI: 10.1371/journal.pone.0234772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 06/02/2020] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma is the most aggressive primary brain tumor leading to death in most of patients. It comprises almost 50-55% of all gliomas with an incidence rate of 2-3 per 100,000. Despite its rarity, overall mortality of glioblastoma is comparable to the most frequent tumors. The current standard treatment combines surgical resection, radiotherapy and chemotherapy with temozolomide. In spite of this aggressive multimodality protocol, prognosis of glioblastoma is poor and the median survival remains about 12-14.5 months. In this regard, new therapeutic approaches should be developed to improve the life quality and survival time of the patient after the initial diagnosis. Before switching to clinical trials in humans, all innovative therapeutic methods must be studied first on a relevant animal model in preclinical settings. In this regard, we validated the feasibility of intratumoral delivery of a holmium (Ho) microparticle suspension to an induced U87 glioblastoma model. Among the different radioactive beta emitters, 166Ho emits high-energy β(-) radiation and low-energy γ radiation. β(-) radiation is an effective means for tumor destruction and γ rays are well suited for imaging (SPECT) and consequent dosimetry. In addition, the paramagnetic Ho nucleus is a good asset to perform MRI imaging. In this study, five minipigs, implanted with our glioblastoma model were used to test the injectability of 165Ho (stable) using a bespoke injector and needle. The suspension was produced in the form of Ho microparticles and injected inside the tumor by a technique known as microbrachytherapy using a stereotactic system. At the end of this trial, it was found that the 165Ho suspension can be injected successfully inside the tumor with absence or minimal traces of Ho reflux after the injections. This injection technique and the use of the 165Ho suspension needs to be further assessed with radioactive 166Ho in future studies.
Collapse
Affiliation(s)
- Mehrdad Khoshnevis
- ICE (Interactions Cellules Environnement), UPSP 2016.A104, VetAgro Sup, University of Lyon1, Marcy l’Etoile, France
| | - Claude Carozzo
- ICE (Interactions Cellules Environnement), UPSP 2016.A104, VetAgro Sup, University of Lyon1, Marcy l’Etoile, France
| | | | | | - Catherine Bonnefont-Rebeix
- ICE (Interactions Cellules Environnement), UPSP 2016.A104, VetAgro Sup, University of Lyon1, Marcy l’Etoile, France
| | - Sara Belluco
- ICE (Interactions Cellules Environnement), UPSP 2016.A104, VetAgro Sup, University of Lyon1, Marcy l’Etoile, France
| | | | - Lionel Marcon
- Institut Lumière Matière, UMR CNRS 5306, UCBL, Campus LyonTech—La Doua, Villeurbanne, France
| | - Olivier Tillement
- Institut Lumière Matière, UMR CNRS 5306, UCBL, Campus LyonTech—La Doua, Villeurbanne, France
| | | | | | - Ilyes Zahi
- Advanced Accelerator Applications, Saint-Genis Pouilly, France
| | - Thierry Buronfosse
- Department of Endocrinology, VetAgro Sup, University of Lyon1, Marcy l’Etoile, France
| | - Thierry Roger
- ICE (Interactions Cellules Environnement), UPSP 2016.A104, VetAgro Sup, University of Lyon1, Marcy l’Etoile, France
| | - Frédérique Ponce
- ICE (Interactions Cellules Environnement), UPSP 2016.A104, VetAgro Sup, University of Lyon1, Marcy l’Etoile, France
- Clinical Oncology Unit, VetAgro Sup, University of Lyon1, Marcy l’Etoile, France
| |
Collapse
|
79
|
Zheng B, Chen T. MiR-489-3p inhibits cell proliferation, migration, and invasion, and induces apoptosis, by targeting the BDNF-mediated PI3K/AKT pathway in glioblastoma. Open Life Sci 2020; 15:274-283. [PMID: 33817216 PMCID: PMC7874546 DOI: 10.1515/biol-2020-0024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/09/2019] [Accepted: 01/03/2020] [Indexed: 12/15/2022] Open
Abstract
Among astrocyte tumors, glioblastoma (GBM) is the most malignant glioma, highly aggressive and invasive, with extremely poor prognosis. Previous research has reported that microRNAs (miRNAs) participate in the progression of many cancers. Thus, this study aimed to explore the role and the underlying mechanisms of microRNA (miR)-489-3p in GBM progression. The expression of miR-489-3p and brain-derived neurotrophic factor (BDNF) mRNA was measured by quantitative real-time polymerase chain reaction. Western blot analysis was used to detect BDNF protein and the PI3K/AKT pathway-related protein. Cell proliferation, apoptosis, migration, and invasion were analyzed using CKK-8 assay, flow cytometry, and transwell assay, respectively. The interaction between BDNF and miR-489-3p was explored by luciferase reporter assay and RNA immunoprecipitation (RIP) assay. MiR-489-3p was down-regulated and BDNF was up-regulated in GBM tissues and cells. MiR-489-3p re-expression or BDNF knockdown inhibited GBM cell proliferation, migration, and invasion, and promoted apoptosis. BDNF was a target of miR-489-3p, and BDNF up-regulation reversed the effects of miR-489-3p on GBM cells. The protein levels of p-AKT and p-PI3K were notably reduced in GBM cells by overexpression of miR-489-3p, but were rescued following BDNF up-regulation. Therefore, miR-489-3p inhibited proliferation, migration, and invasion, and induced apoptosis, by targeting the BDNF-mediated PI3K/AKT pathway in GBM, providing new strategies for clinical treatment of GBM.
Collapse
Affiliation(s)
- Bo Zheng
- Department of Neurosurgery, Jingzhou Central Hospital, Hubei Province, Jingzhou, 434020, China
| | - Tao Chen
- Department of Neurosurgery, Jingzhou Central Hospital, Hubei Province, Jingzhou, 434020, China
| |
Collapse
|
80
|
Khan L, Soliman H, Sahgal A, Perry J, Xu W, Tsao MN. External beam radiation dose escalation for high grade glioma. Cochrane Database Syst Rev 2020; 5:CD011475. [PMID: 32437039 PMCID: PMC7389526 DOI: 10.1002/14651858.cd011475.pub3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND This is an updated version of the original Cochrane Review published in Issue 8, 2016. High grade glioma (HGG) is a rapidly growing brain tumour in the supporting cells of the nervous system, with several subtypes such as glioblastoma (grade IV astrocytoma), anaplastic (grade III) astrocytoma and anaplastic (grade III) oligodendroglioma. Studies have investigated the best strategy to give radiation to people with HGG. Conventional fractionated radiotherapy involves giving a daily radiation dose (called a fraction) of 180 cGy to 200 cGy. Hypofractionated radiotherapy uses higher daily doses, which reduces the overall number of fractions and treatment time. Hyperfractionated radiotherapy which uses a lower daily dose with a greater number of fractions and multiple fractions per day to deliver a total dose at least equivalent to external beam daily conventionally fractionated radiotherapy in the same time frame. The aim is to reduce the potential for late toxicity. Accelerated radiotherapy (dose escalation) refers to the delivery of multiple fractions per day using daily doses of radiation consistent with external beam daily conventionally fractionated radiotherapy doses. The aim is to reduce the overall treatment time; typically, two or three fractions per day may be delivered with a six to eight hour gap between fractions. OBJECTIVES To assess the effects of postoperative external beam radiation dose escalation in adults with HGG. SEARCH METHODS We searched CENTRAL, MEDLINE Ovid and Embase Ovid to August 2019 for relevant randomised phase III trials. SELECTION CRITERIA We included adults with a pathological diagnosis of HGG randomised to the following external beam radiation regimens: daily conventionally fractionated radiotherapy versus no radiotherapy; hypofractionated radiotherapy versus daily conventionally fractionated radiotherapy; hyperfractionated radiotherapy versus daily conventionally fractionated radiotherapy or accelerated radiotherapy versus daily conventionally fractionated radiotherapy. DATA COLLECTION AND ANALYSIS The primary outcomes were overall survival and adverse effects. The secondary outcomes were progression free survival and quality of life. We used the standard methodological procedures expected by Cochrane. We assessed the certainty of the evidence using the GRADE approach. MAIN RESULTS Since the last version of this review, we identified no new relevant trials for inclusion. We included 11 randomised controlled trials (RCTs) with 2062 participants and 1537 in the relevant arms for this review. There was an overall survival benefit for people with HGG receiving postoperative radiotherapy compared to the participants receiving postoperative supportive care. For the four pooled RCTs (397 participants), the overall hazard ratio (HR) for survival was 2.01 favouring postoperative radiotherapy (95% confidence interval (CI) 1.58 to 2.55; P < 0.00001; moderate-certainty evidence). Although these trials may not have completely reported adverse effects, they did not note any significant toxicity attributable to radiation. Progression free survival and quality of life could not be pooled due to lack of data. Overall survival was similar between hypofractionated and conventional radiotherapy in five trials (943 participants), where the HR was 0.95 (95% CI 0.78 to 1.17; P = 0.63; very low-certainty evidence. The trials reported that hypofractionated and conventional radiotherapy were well tolerated with mild acute adverse effects. These trials only reported one participant in the hypofractionated arm developing symptomatic radiation necrosis that required surgery. Progression free survival and quality of life could not be pooled due to the lack of data. Overall survival was similar between hypofractionated and conventional radiotherapy in the subset of two trials (293 participants) which included participants aged 60 years and older with glioblastoma. For this category, the HR was 1.16 (95% CI 0.92 to 1.46; P = 0.21; high-certainty evidence). There were two trials which compared hyperfractionated radiotherapy versus conventional radiation and one trial which compared accelerated radiotherapy versus conventional radiation. However, the results could not be pooled. The conventionally fractionated radiotherapy regimens were 4500 cGy to 6000 cGy given in 180 cGy to 200 cGy daily fractions, over five to six weeks. All trials generally included participants with World Health Organization (WHO) performance status from 0 to 2 and Karnofsky performance status of 50 and higher. The risk of selection bias was generally low among these RCTs. The number of participants lost to follow-up for the outcome of overall survival was low. Attrition, performance, detection and reporting bias for the outcome of overall survival was low. There was unclear attrition, performance, detection and reporting bias relating to the outcomes of adverse effects, progression free survival and quality of life. AUTHORS' CONCLUSIONS Postoperative conventional daily radiotherapy probably improves survival for adults with good performance status and HGG compared to no postoperative radiotherapy. Hypofractionated radiotherapy has similar efficacy for survival compared to conventional radiotherapy, particularly for individuals aged 60 years and older with glioblastoma. There are insufficient data regarding hyperfractionation versus conventionally fractionated radiation (without chemotherapy) and for accelerated radiation versus conventionally fractionated radiation (without chemotherapy). There are HGG subsets who have poor prognosis even with treatment (e.g. glioblastoma histology, older age and poor performance status). These HGG individuals with poor prognosis have generally been excluded from randomised trials based on poor performance status. No randomised trial has compared comfort measures or best supportive care with an active intervention using radiotherapy or chemotherapy in these people with poor prognosis. Since the last version of this review, we found no new relevant studies. The search identified three new trials, but all were excluded as none had a conventionally fractionated radiotherapy arm.
Collapse
Affiliation(s)
- Luluel Khan
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Hany Soliman
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - James Perry
- Crolla Endowed Chair of Neuro-Oncology Research, Sunnybrook Health Sciences Centre and Odette Cancer Centre, Toronto, Canada
| | - Wei Xu
- Department of Biostatistics, University of Toronto, Toronto, Canada
| | - May N Tsao
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| |
Collapse
|
81
|
Bhat K, Saki M, Vlashi E, Cheng F, Duhachek-Muggy S, Alli C, Yu G, Medina P, He L, Damoiseaux R, Pellegrini M, Zemke NR, Nghiemphu PL, Cloughesy TF, Liau LM, Kornblum HI, Pajonk F. The dopamine receptor antagonist trifluoperazine prevents phenotype conversion and improves survival in mouse models of glioblastoma. Proc Natl Acad Sci U S A 2020; 117:11085-11096. [PMID: 32358191 PMCID: PMC7245100 DOI: 10.1073/pnas.1920154117] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the deadliest adult brain cancer, and all patients ultimately succumb to the disease. Radiation therapy (RT) provides survival benefit of 6 mo over surgery alone, but these results have not improved in decades. We report that radiation induces a glioma-initiating cell phenotype, and we have identified trifluoperazine (TFP) as a compound that interferes with this phenotype conversion. TFP causes loss of radiation-induced Nanog mRNA expression, and activation of GSK3 with consecutive posttranslational reduction in p-Akt, Sox2, and β-catenin protein levels. TFP did not alter the intrinsic radiation sensitivity of glioma-initiating cells (GICs). Continuous treatment with TFP and a single dose of radiation reduced the number of GICs in vivo and prolonged survival in syngeneic and patient-derived orthotopic xenograft (PDOX) mouse models of GBM. Our findings suggest that the combination of a dopamine receptor antagonist with radiation enhances the efficacy of RT in GBM by preventing radiation-induced phenotype conversion of radiosensitive non-GICs into treatment-resistant, induced GICs (iGICs).
Collapse
Affiliation(s)
- Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Mohammad Saki
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| | - Fei Cheng
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Sara Duhachek-Muggy
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Claudia Alli
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Garrett Yu
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Paul Medina
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Ling He
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Robert Damoiseaux
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Molecular Screening Shared Resource, University of California, Los Angeles, CA 90095
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095
| | - Nathan R Zemke
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Phioanh Leia Nghiemphu
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Neurology, University of California, Los Angeles, CA 90095
| | - Timothy F Cloughesy
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Neurology, University of California, Los Angeles, CA 90095
| | - Linda M Liau
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Neurosurgery, University of California, Los Angeles, CA 90095
| | - Harley I Kornblum
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Neuropsychiatric Institute-Semel Institute for Neuroscience & Human Behavior, University of California, Los Angeles, CA 90095
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095;
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| |
Collapse
|
82
|
Ruff M, Kizilbash S, Buckner J. Further understanding of glioma mechanisms of pathogenesis: implications for therapeutic development. Expert Rev Anticancer Ther 2020; 20:355-363. [PMID: 32301635 DOI: 10.1080/14737140.2020.1757440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Recent discoveries in the molecular makeup of gliomas, the relationship of certain molecular drivers, and the patient's response to therapy and overall prognosis have resulted in a paradigm shift and redefined our understanding of glioma and revealed potential vulnerabilities within this recalcitrant and lethal disease.Areas covered: We summarize the current classification of malignant glioma in the context of the historical background, current data-driven treatment strategies, and recent discoveries of the mechanisms of pathogenesis of this disease which recapitulates the developing brain. We describe the relationship to common genetic alterations found in glioma, and possible avenues to exploit these newly revealed mechanisms.Expert opinion: Improved understanding of the molecular underpinnings of this disease has been directly translated into treatment decisions and an improved ability to counsel patients regarding their prognosis. We are beginning to see the first glimmer of a return on the investment in regard to immunotherapy in malignant glioma, with further anticipated successful exploitations of the unique pathophysiology of glioma.
Collapse
Affiliation(s)
- Michael Ruff
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Sani Kizilbash
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jan Buckner
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
83
|
Kirstein A, Schmid TE, Combs SE. The Role of miRNA for the Treatment of MGMT Unmethylated Glioblastoma Multiforme. Cancers (Basel) 2020; 12:cancers12051099. [PMID: 32354046 PMCID: PMC7281574 DOI: 10.3390/cancers12051099] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common high-grade intracranial tumor in adults. It is characterized by uncontrolled proliferation, diffuse infiltration due to high invasive and migratory capacities, as well as intense resistance to chemo- and radiotherapy. With a five-year survival of less than 3% and an average survival rate of 12 months after diagnosis, GBM has become a focus of current research to urgently develop new therapeutic approaches in order to prolong survival of GBM patients. The methylation status of the promoter region of the O6-methylguanine–DNA methyltransferase (MGMT) is nowadays routinely analyzed since a methylated promoter region is beneficial for an effective response to temozolomide-based chemotherapy. Furthermore, several miRNAs were identified regulating MGMT expression, apart from promoter methylation, by degrading MGMT mRNA before protein translation. These miRNAs could be a promising innovative treatment approach to enhance Temozolomide (TMZ) sensitivity in MGMT unmethylated patients and to increase progression-free survival as well as long-term survival. In this review, the relevant miRNAs are systematically reviewed.
Collapse
Affiliation(s)
- Anna Kirstein
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Thomas E. Schmid
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Stephanie E. Combs
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-4501
| |
Collapse
|
84
|
Mohammadi AM, Sharma M, Beaumont TL, Juarez KO, Kemeny H, Dechant C, Seas A, Sarmey N, Lee BS, Jia X, Fecci PE, Baehring J, Moliterno J, Chiang VL, Ahluwalia MS, Kim AH, Barnett GH, Leuthardt EC. Upfront Magnetic Resonance Imaging-Guided Stereotactic Laser-Ablation in Newly Diagnosed Glioblastoma: A Multicenter Review of Survival Outcomes Compared to a Matched Cohort of Biopsy-Only Patients. Neurosurgery 2020; 85:762-772. [PMID: 30476325 DOI: 10.1093/neuros/nyy449] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/21/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Laser ablation (LA) is used as an upfront treatment in patients with deep seated newly diagnosed Glioblastoma (nGBM). OBJECTIVE To evaluate the outcomes of LA in patients with nGBM and compare them with a matched biopsy-only cohort. METHODS Twenty-four nGBM patients underwent upfront LA at Cleveland clinic, Washington University in St. Louis, and Yale University (6/2011-12/2014) followed by chemo/radiotherapy. Also, 24 out of 171 nGBM patients with biopsy followed by chemo/radiotherapy were matched based on age (< 70 vs ≥ 70), gender, tumor location (deep vs lobar), and volume (<11 cc vs ≥11 cc). Progression-free survival (PFS), overall survival (OS), and disease-specific PFS and OS were outcome measures. Three prognostic groups were identified based on extent of tumor ablation by thermal-damage-threshold (TDT)-lines. RESULTS The median tumor volume in LA (n = 24) and biopsy only (n = 24) groups was 9.3 cm3 and 8.2 cm3 respectively. Overall, median estimate of OS and PFS in LA cohort was 14.4 and 4.3 mo compared to 15.8 mo and 5.9 mo for biopsy only cohort. On multivariate analysis, favorable TDT-line prognostic groups were associated with lower incidence of disease specific death (P = .03) and progression (P = .05) compared to other groups including biopsy only cohort. Only age (<70 yr, P = .02) and tumor volume (<11 cc, P = .03) were favorable prognostic factors for OS. CONCLUSION The maximum tumor coverage by LA followed by radiation/chemotherapy is an effective treatment modality in patients with nGBM, compared to biopsy only cohort. The TDT-line prognostic groups were independent predictor of disease specific death and progression after LA.
Collapse
Affiliation(s)
- Alireza M Mohammadi
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Mayur Sharma
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Thomas L Beaumont
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Kevin O Juarez
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Hanna Kemeny
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Cosette Dechant
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Andreas Seas
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Nehaw Sarmey
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Bryan S Lee
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Xuefei Jia
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio
| | - Peter E Fecci
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Joachim Baehring
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Jennifer Moliterno
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Veronica L Chiang
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Manmeet S Ahluwalia
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Albert H Kim
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gene H Barnett
- The Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Eric C Leuthardt
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Center for Innovation in Neuroscience and Technology, Washington University School of Medicine, St. Louis, Missouri
- Department of Mechanical Engineering and Material Science, Center for Innovation in Neuroscience and Technology, Washington University, School of Medicine, St. Louis, Missouri
| |
Collapse
|
85
|
Zhao YY, Wan QS, Hao Z, Zhu HX, Xing ZL, Li MH. Clinical nomogram for predicting the survival of patients with cerebral anaplastic gliomas. Medicine (Baltimore) 2020; 99:e19416. [PMID: 32150092 PMCID: PMC7478695 DOI: 10.1097/md.0000000000019416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The present study aimed to develop an effective nomogram for predicting the overall survival (OS) of patients with cerebral anaplastic glioma (AG).This study included 1939 patients diagnosed with AG between 1973 and 2013 who were identified using the Surveillance, Epidemiology, and End Results database. A multivariate Cox regression analysis revealed that age, histology, tumor site, marital status, radiotherapy, and surgery were independent prognostic factors and, thus, these factors were selected to build a clinical nomogram. Harrell's concordance index (C-index) and a calibration curve were formulated to evaluate the discrimination and calibration of the nomogram using bootstrapping.A nomogram was developed to predict 5- and 9-year OS rates based on 6 independent prognostic factors identified in the training set: age, tumor site, marital status, histology, radiotherapy, and surgery (P < .05). The Harrell's concordance index values of the training and validation sets were 0.776 (0.759-0.793) and 0.766 (0.739-0.792), respectively. The calibration curve exhibited good consistency with the actual observation curve in both sets.Although the prognostic value of the World Health Organization (WHO) classification has been validated, we developed a novel nomogram based on readily available clinical variables in terms of demographic data, therapeutic modalities, and tumor characteristics to predict the survival of AG patients. When used in combination with the WHO classification system, this clinical nomogram can aid clinicians in making individualized predictions of AG patient survival and improving treatment strategies.
Collapse
Affiliation(s)
| | - Qin-Si Wan
- Department of gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | | | | | | | | |
Collapse
|
86
|
Potez M, Bouchet A, Flaender M, Rome C, Collomb N, Grotzer M, Krisch M, Djonov V, Balosso J, Brun E, Laissue JA, Serduc R. Synchrotron X-Ray Boost Delivered by Microbeam Radiation Therapy After Conventional X-Ray Therapy Fractionated in Time Improves F98 Glioma Control. Int J Radiat Oncol Biol Phys 2020; 107:360-369. [PMID: 32088292 DOI: 10.1016/j.ijrobp.2020.02.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/29/2020] [Accepted: 02/02/2020] [Indexed: 10/25/2022]
Abstract
PURPOSE Synchrotron microbeam radiation therapy (MRT) is based on the spatial fractionation of the incident, highly collimated synchrotron beam into arrays of parallel microbeams depositing several hundred grays. It appears relevant to combine MRT with a conventional treatment course, preparing a treatment scheme for future patients in clinical trials. The efficiency of MRT delivered after several broad-beam (BB) fractions to palliate F98 brain tumors in rats in comparison with BB fractions alone was evaluated in this study. METHODS AND MATERIALS Rats bearing 106 F98 cells implanted in the caudate nucleus were irradiated by 5 fractions in BB mode (3 × 6 Gy + 2 × 8 Gy BB) or by 2 boost fractions in MRT mode to a total of 5 fractions (3 × 6 Gy BB + MRT 2 × 8 Gy valley dose; peak dose 181 Gy [50/200 μm]). Tumor growth was evaluated in vivo by magnetic resonance imaging follow-up at T-1, T7, T12, T15, T20, and T25 days after radiation therapy and by histology and flow cytometry. RESULTS MRT-boosted tumors displayed lower cell density and cell proliferation compared with BB-irradiated tumors. The MRT boost completely stopped tumor growth during ∼4 weeks and led to a significant increase in median survival time, whereas tumors treated with BB alone recurred within a few days after the last radiation fraction. CONCLUSIONS The first evidence is presented that MRT, delivered as a boost of conventionally fractionated irradiation by orthovoltage broad x-ray beams, is feasible and more efficient than conventional radiation therapy alone.
Collapse
Affiliation(s)
- Marine Potez
- Inserm UA7, Rayonnement synchrotron pour la recherche médicale (STROBE), Université Grenoble Alpes, Grenoble, France
| | - Audrey Bouchet
- Inserm UA7, Rayonnement synchrotron pour la recherche médicale (STROBE), Université Grenoble Alpes, Grenoble, France; Institute of Anatomy, Group Tomographic and Clinical Anatomy, University of Bern, Bern, Switzerland.
| | - Mélanie Flaender
- Inserm UA7, Rayonnement synchrotron pour la recherche médicale (STROBE), Université Grenoble Alpes, Grenoble, France
| | - Claire Rome
- Team Functional NeuroImaging and Brain Perfusion, Inserm, France; Grenoble Institut des Neurosciences, Université Grenoble Alpes, La Tronche, France
| | - Nora Collomb
- Team Functional NeuroImaging and Brain Perfusion, Inserm, France; Grenoble Institut des Neurosciences, Université Grenoble Alpes, La Tronche, France
| | - Michael Grotzer
- Department of Oncology, University Children's Hospital of Zurich, Switzerland
| | - Michael Krisch
- European Synchrotron Radiation Facility, Grenoble, France
| | - Valentin Djonov
- Institute of Anatomy, Group Tomographic and Clinical Anatomy, University of Bern, Bern, Switzerland
| | - Jacques Balosso
- Inserm UA7, Rayonnement synchrotron pour la recherche médicale (STROBE), Université Grenoble Alpes, Grenoble, France
| | - Emmanuel Brun
- Inserm UA7, Rayonnement synchrotron pour la recherche médicale (STROBE), Université Grenoble Alpes, Grenoble, France
| | | | - Raphaël Serduc
- Inserm UA7, Rayonnement synchrotron pour la recherche médicale (STROBE), Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
87
|
Kim HS, Kwon SL, Choi SH, Hwang I, Kim TM, Park CK, Park SH, Won JK, Kim IH, Lee ST. Prognostication of anaplastic astrocytoma patients: application of contrast leakage information of dynamic susceptibility contrast-enhanced MRI and dynamic contrast-enhanced MRI. Eur Radiol 2020; 30:2171-2181. [PMID: 31953664 DOI: 10.1007/s00330-019-06598-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/13/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE To examine the applicability of contrast leakage information from dynamic susceptibility contrast-enhanced (DSC) MRI and dynamic contrast-enhanced (DCE) MRI to determine which one is the most valuable surrogate imaging biomarker for predicting disease progression in anaplastic astrocytoma (AA) patients. MATERIALS AND METHODS This study was approved by the institutional review board (IRB), which waived informed consent. A total of seventy-three AA patients who had undergone preoperative DCE and DSC MRI and received standard treatment, including partial resection or biopsy followed by radiation therapy, were included in this retrospective study. Based on Response Assessment in Neuro-Oncology (RANO), patients were sorted into progression (n = 21) and non-progression (n = 52) groups. Tumor boundaries were defined as high-signal intensity (SI) lesions on fluid-attenuated inversion recovery (FLAIR) imaging, where we analyzed mean pharmacokinetic parameters (Ktrans, Vp, and Ve) from DCE MRI and contrast leakage information (mean extraction fraction (EF)) from DSC MRI. RESULTS Mean Ve and mean EF were significantly higher in patients with progression-free survival (PFS) < 18 months than in those with PFS ≥ 18 months. For distinguishing the group with PFS < 18 months, AUC values were calculated using the mean Ve value (AUC = 0.716). The Kaplan-Meier survival analysis revealed that mean Ve value was significantly correlated with PFS. In Cox proportional-hazards regression, only the mean Ve value was found to be significantly associated with PFS. CONCLUSION We found that the mean Ve value based on high-SI tumor lesions on FLAIR imaging was capable of predicting outcomes of AA patients as a potential surrogate imaging biomarker. KEY POINTS • Mean Ve(2.152 ± 1.857 vs. 1.173 ± 1.408) was significantly higher in anaplastic astrocytoma patients with PFS < 18 months that in those with PFS ≥ 18 months (p = 0.02). • Cox proportional-hazards regression showed that only mean Ve(p = 0.034) was significantly associated with PFS, regardless of IDH mutation status, in anaplastic astrocytoma patients.
Collapse
Affiliation(s)
- Hee Soo Kim
- College of Medicine, Seoul National University, Seoul, South Korea
| | - Se Lee Kwon
- College of Medicine, Seoul National University, Seoul, South Korea
| | - Seung Hong Choi
- Department of Radiology, College of Medicine, Seoul National University, 28, Yongon-dong, Chongno-gu, Seoul, 110-744, South Korea.
- Center for Nanoparticle Research, Institute for Basic Science, Seoul, South Korea.
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 151-742, South Korea.
| | - Inpyeong Hwang
- Department of Radiology, College of Medicine, Seoul National University, 28, Yongon-dong, Chongno-gu, Seoul, 110-744, South Korea
- Center for Nanoparticle Research, Institute for Basic Science, Seoul, South Korea
| | - Tae Min Kim
- Department of Internal Medicine, Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Biomedical Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
| | - Sung-Hye Park
- Department of Pathology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jae-Kyung Won
- Department of Pathology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Il Han Kim
- Department of Radiation Oncology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
| | - Soon Tae Lee
- Department of Neurology, College of Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
88
|
Thomas RP, Nagpal S, Iv M, Soltys SG, Bertrand S, Pelpola JS, Ball R, Yang J, Sundaram V, Lavezo J, Born D, Vogel H, Brown JM, Recht LD. Macrophage Exclusion after Radiation Therapy (MERT): A First in Human Phase I/II Trial using a CXCR4 Inhibitor in Glioblastoma. Clin Cancer Res 2019; 25:6948-6957. [PMID: 31537527 PMCID: PMC6891194 DOI: 10.1158/1078-0432.ccr-19-1421] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/17/2019] [Accepted: 09/11/2019] [Indexed: 01/18/2023]
Abstract
PURPOSE Preclinical studies have demonstrated that postirradiation tumor revascularization is dependent on a stromal cell-derived factor-1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR4)-driven process in which myeloid cells are recruited from bone marrow. Blocking this axis results in survival improvement in preclinical models of solid tumors, including glioblastoma (GBM). We conducted a phase I/II study to determine the safety and efficacy of Macrophage Exclusion after Radiation Therapy (MERT) using the reversible CXCR4 inhibitor plerixafor in patients with newly diagnosed glioblastoma. PATIENTS AND METHODS We enrolled nine patients in the phase I study and an additional 20 patients in phase II using a modified toxicity probability interval (mTPI) design. Plerixafor was continuously infused intravenously via a peripherally inserted central catheter (PICC) line for 4 consecutive weeks beginning at day 35 of conventional treatment with concurrent chemoradiation. Blood serum samples were obtained for pharmacokinetic analysis. Additional studies included relative cerebral blood volume (rCBV) analysis using MRI and histopathology analysis of recurrent tumors. RESULTS Plerixafor was well tolerated with no drug-attributable grade 3 toxicities observed. At the maximum dose of 400 μg/kg/day, biomarker analysis found suprathreshold plerixafor serum levels and an increase in plasma SDF-1 levels. Median overall survival was 21.3 months [95% confidence interval (CI), 15.9-NA] with a progression-free survival of 14.5 months (95% CI, 11.9-NA). MRI and histopathology support the mechanism of action to inhibit postirradiation tumor revascularization. CONCLUSIONS Infusion of the CXCR4 inhibitor plerixafor was well tolerated as an adjunct to standard chemoirradiation in patients with newly diagnosed GBM and improves local control of tumor recurrences.
Collapse
Affiliation(s)
- Reena P Thomas
- Department of Neurology, Division of Neuro Oncology, Stanford, California.
| | - Seema Nagpal
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| | - Michael Iv
- Department of Radiology, Division of Neuro Radiology, Stanford, California
| | | | - Sophie Bertrand
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| | - Judith S Pelpola
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| | - Robyn Ball
- Department of Medicine, Quantitative Sciences Unit, Stanford, California
| | - Jaden Yang
- Department of Medicine, Quantitative Sciences Unit, Stanford, California
| | - Vandana Sundaram
- Department of Medicine, Quantitative Sciences Unit, Stanford, California
| | - Jonathan Lavezo
- Department of Pathology, Division of Neuro Pathology, Stanford University, Stanford, California
| | - Donald Born
- Department of Pathology, Division of Neuro Pathology, Stanford University, Stanford, California
| | - Hannes Vogel
- Department of Pathology, Division of Neuro Pathology, Stanford University, Stanford, California
| | - J Martin Brown
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| | - Lawrence D Recht
- Department of Neurology, Division of Neuro Oncology, Stanford, California
| |
Collapse
|
89
|
Mathen P, Rowe L, Mackey M, Smart D, Tofilon P, Camphausen K. Radiosensitizers in the temozolomide era for newly diagnosed glioblastoma. Neurooncol Pract 2019; 7:268-276. [PMID: 32537176 DOI: 10.1093/nop/npz057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a challenging diagnosis with almost universally poor prognosis. Though the survival advantage of postoperative radiation (RT) is well established, around 90% of patients will fail in the RT field. The high likelihood of local failure suggests the efficacy of RT needs to be improved to improve clinical outcomes. Radiosensitizers are an established method of enhancing RT cell killing through the addition of a pharmaceutical agent. Though the majority of trials using radiosensitizers have historically been unsuccessful, there continues to be interest with a variety of approaches having been employed. Epidermal growth factor receptor inhibitors, histone deacetylase inhibitors, antiangiogenic agents, and a number of other molecularly targeted agents have all been investigated as potential methods of radiosensitization in the temozolomide era. Outcomes have varied both in terms of toxicity and survival, but some agents such as valproic acid and bortezomib have demonstrated promising results. However, reporting of results in phase 2 trials in newly diagnosed GBM have been inconsistent, with no standard in reporting progression-free survival and toxicity. There is a pressing need for investigation of new agents; however, nearly all phase 3 trials of GBM patients of the past 25 years have demonstrated no improvement in outcomes. One proposed explanation for this is the selection of agents lacking sufficient preclinical data and/or based on poorly designed phase 2 trials. Radiosensitization may represent a viable strategy for improving GBM outcomes in newly diagnosed patients, and further investigation using agents with promising phase 2 data is warranted.
Collapse
Affiliation(s)
- Peter Mathen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Lindsay Rowe
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Megan Mackey
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - DeeDee Smart
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Philip Tofilon
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
90
|
Saki M, Bhat K, Sodhi SS, Nguyen NT, Kornblum HI, Pajonk F. Effects of Brain Irradiation in Immune-Competent and Immune-Compromised Mouse Models. Radiat Res 2019; 193:186-194. [PMID: 31774721 DOI: 10.1667/rr15373.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Patient-derived orthotopic xenografts (PDOXs) closely recapitulate primary human glioblastoma (GBM) tumors in terms of histology and genotype. Compared to other mouse strains, NOD-scid IL2Rgammanull (NSG) mice show excellent tumor take rates, which makes them an ideal host for PDOXs. However, NSG mice harbor a mutation in the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs), which renders them relatively radiosensitive. This has been a frequently voiced concern in studies involving ionizing radiation. In this study, we assessed brain toxicity in NSG mice compared to three other different mouse strains frequently used in radiation studies at radiation doses commonly used in experimental combination therapy studies. C3H/Sed/Kam, C57Bl/6, nude and NOD-scid IL2Rgammanull mice received a single dose of 4 Gy to the right brain hemispheres using an image-guided small animal irradiator. Brains were stained using H&E, luxol fast blue, and antibodies against IBA1 and GFAP one, two, four or six months postirradiation. Additional animals of all four strains were exposed to five daily fractions of 2 Gy (5 × 2 Gy), and tissue sections were stained 72 h later against gH2AX, NeuN, GFAP and IBA1. None of the mouse strains displayed radiation-induced toxicity at any of the time points tested. Radiation doses relevant for testing combination therapies can be safely applied to the brains of NSG mice without the occurrence of radiation-induced normal tissue toxicity.
Collapse
Affiliation(s)
- Mohammad Saki
- Departments of Radiation Oncology, David Geffen School of Medicine
| | - Kruttika Bhat
- Departments of Radiation Oncology, David Geffen School of Medicine
| | - Sirajbir S Sodhi
- Departments of Radiation Oncology, David Geffen School of Medicine
| | - Nhan T Nguyen
- Departments of Radiation Oncology, David Geffen School of Medicine
| | - Harley I Kornblum
- Departments of Molecular and Medical Pharmacology.,Departments of Neurology.,Departments of Psychiatry and Biobehavioral Sciences.,Departments of NPI-The Jane and Terry Semel Institute for Neuroscience and Human Behavior.,Departments of Molecular Biology Institute.,Departments of Jonsson Comprehensive Cancer Center at UCLA, University of California, Los Angeles, Los Angeles, California.,Departments of Eli & Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California
| | - Frank Pajonk
- Departments of Radiation Oncology, David Geffen School of Medicine.,Departments of Jonsson Comprehensive Cancer Center at UCLA, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
91
|
Computational studies of anti-cancer drug mediated by graphene and reaction mechanism of drug generated alkyl radical with guanine. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2019.06.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
92
|
Blumenthal DT, Won M, Mehta MP, Gilbert MR, Brown PD, Bokstein F, Brachman DG, Werner-Wasik M, Hunter GK, Valeinis E, Hopkins K, Souhami L, Howard SP, Lieberman FS, Shrieve DC, Wendland MM, Robinson CG, Zhang P, Corn BW. Short delay in initiation of radiotherapy for patients with glioblastoma-effect of concurrent chemotherapy: a secondary analysis from the NRG Oncology/Radiation Therapy Oncology Group database. Neuro Oncol 2019; 20:966-974. [PMID: 29462493 DOI: 10.1093/neuonc/noy017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background We previously reported the unexpected finding of significantly improved survival for newly diagnosed glioblastoma in patients when radiation therapy (RT) was initiated later (>4 wk post-op) compared with earlier (≤2 wk post-op). In that analysis, data were analyzed from 2855 patients from 16 NRG Oncology/Radiotherapy Oncology Group (RTOG) trials conducted prior to the era of concurrent temozolomide (TMZ) with RT. We now report on 1395 newly diagnosed glioblastomas from 2 studies, treated with RT and concurrent TMZ followed by adjuvant TMZ. Our hypothesis was that concurrent TMZ has a synergistic/radiosensitizing mechanism, making RT timing less significant. Methods Data from patients treated with TMZ-based chemoradiation from NRG Oncology/RTOG 0525 and 0825 were analyzed. An analysis comparable to our prior study was performed to determine whether there was still an impact on survival by delaying RT. Overall survival (OS) was investigated using the Kaplan-Meier method and Cox proportional hazards model. Early progression (during time of diagnosis to 30 days after RT completion) was analyzed using the chi-square test. Results Given the small number of patients who started RT early following surgery, comparisons were made between >4 and ≤4 weeks delay of radiation from time of operation. There was no statistically significant difference in OS (hazard ratio = 0.93; P = 0.29; 95% CI: 0.80-1.07) after adjusting for known prognostic factors (recursive partitioning analysis and O6-methylguanine-DNA methyltransferase methylation status). Similarly, the rate of early progression did not differ significantly (P = 0.63). Conclusions We did not observe a significant prognostic influence of delaying radiation when given concurrently with TMZ for newly diagnosed glioblastoma. The effects of early (1-3 wk post-op) or late (>5 wk) initiation of radiation tested in our prior study could not be replicated.
Collapse
Affiliation(s)
| | - Minhee Won
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania, USA
| | | | - Mark R Gilbert
- National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Paul D Brown
- University of Texas-MD Anderson Cancer Center, Houston, Texas, USA.,Mayo Clinic, Rochester, Minnesota, USA
| | | | - David G Brachman
- Saint Joseph's Hospital and Medical Center ACCRUALS for Arizona Oncology Services Foundation, Phoenix, Arizona, USA
| | | | | | - Egils Valeinis
- Paulus Stradins Clinical University Hospital-EORTC, Riga, Latvia
| | | | | | | | | | - Dennis C Shrieve
- University of Utah Health Science Center, Salt Lake City, Utah, USA
| | | | | | - Peixin Zhang
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
93
|
Kumar N, Hanumanthappa N, Miriyala R, Vyas S, Salunke P, Oinam AS, Yadav BS, Madan R, Dracham C, Kapoor R. Hurdles in radiation planning for glioblastoma: Can delayed-contrast enhanced computed tomography be a potential solution? Asia Pac J Clin Oncol 2019; 15:e103-e108. [PMID: 30698349 DOI: 10.1111/ajco.13111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/02/2018] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Conformal radiation is the standard of care in treatment of glioblastoma. Although co-registration of magnetic resonance imaging (MRI) with early contrast enhanced computed tomography (CECT) is recommended for target delineation by consensus guidelines, ground realities in developing countries often result in availability of less-than-ideal MR sequences for treatment planning. Purpose of this study is to analyze the impact of incorporation of delayed-CECT sequences for radiation planning in glioblastomas, as an adjunct or alternative to MRI. METHODS Case records of all patients of glioblastoma treated at our center between 2011 and 2014 were retrospectively evaluated. Gross treatment volumes were delineated on T1 contrast MRI (m-GTV), early CECT (e-GTV) and delayed CECT (d-GTV); volumetric comparisons were made using repeated measures analysis of variance and pair-wise analysis. RESULTS Although 96% of registered patients underwent postoperative MRI, only 38% of them had desirable sequences suitable for co-registration. Median duration between acquisition of postoperative MRI and surgery was 45 days (range, 33-60), whereas that between MRI and treatment-planning CT was 5 days (range, 1-10). Statistically significant differences (P < 0.0001) were obtained between mean volumes of e-GTV (41.20cc), d-GTV (58.09cc) and m-GTV (60.52cc). Although the mean GTV increased by 46% between early CECT and MRI, the difference was only 4% between delayed CECT and MRI. CONCLUSION Delayed CECT is superior to early CECT for co-registration with MRI for target delineation, especially when available MR sequences are less-than-ideal for treatment planning, and can be considered as the most appropriate adjunct as well as an alternative to MRI, compared to early CECT.
Collapse
Affiliation(s)
| | | | | | - Sameer Vyas
- Department of Radiodiagnosis, PGIMER, Chandigarh, India
| | | | - Arun S Oinam
- Department of Radiotherapy, PGIMER, Chandigarh, India
| | - Budhi S Yadav
- Department of Radiotherapy, PGIMER, Chandigarh, India
| | - Renu Madan
- Department of Radiotherapy, PGIMER, Chandigarh, India
| | | | - Rakesh Kapoor
- Department of Radiotherapy, PGIMER, Chandigarh, India
| |
Collapse
|
94
|
Klekner A, Tóth J, Virga J, Hortobágyi T, Dér Á, Szemcsák C, Reményi-Puskár J, Bognár L. Influence of Oncotherapy and Clinical Parameters on Survival of Glioblastoma Patients: A Single Center Experience. Neurol India 2019; 67:1066-1073. [PMID: 31512637 DOI: 10.4103/0028-3886.266257] [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] [Indexed: 11/04/2022]
Abstract
Background Routine administration of temozolomide (TMZ) in the treatment protocol of glioblastoma in the last few years resulted in improving survival parameters of these patients but efficacy of supplementary bevacizumab (BVC) monotherapy has not been evidently proven. In this study, the effectiveness of different postoperative therapy for glioblastoma patients treated in our institute was evaluated. In addition, the prognostic value of clinical parameters on survival was also analyzed. Methods Accordance of clinical parameters (age, gender, tumor localization, size, side, Karnofsky performance score, and extension of tumor removal), postoperative treatment (radiotherapy [RT], RT + TMZ, RT + TMZ + BVC), and survival data were tested by 104 patients operated on glioblastoma in the Department of Neurosurgery, University of Debrecen between 2002 and 2012. Results Concurrent chemo-RT resulted in significant longer overall survival (OS) than RT alone (PRTvs.RT + TMZ = 0.0219) and BVC treatment after progression during TMZ also elongated survival significantly (PRT vs. RT + TMZ + BVC < 0.0001; PRT + TMZvs.RT + TMZ + BVC = 0.0022), respectively. Clinical parameters showed no significant influence on OS in comparison with different methods of postoperative oncotherapy. Conclusions Both TMZ and BVC had a beneficial effect on glioblastoma patients' survival, but tested clinical parameters showed no evident accordance with final outcome. Although neurosurgery has an indispensable role in resecting space occupying tumors and providing good postoperative performance score patients for oncotherapy, the survival of glioblastoma patients depends rather on radio- and chemo-sensitivity than tested clinical parameters.
Collapse
Affiliation(s)
- Almos Klekner
- Department of Neurosurgery, University of Debrecen, Clinical Center, Debrecen, Nagyerdei krt. 98, 4032, Hungary
| | - Judit Tóth
- Department of Oncology, University of Debrecen, Clinical Center, Debrecen, Nagyerdei krt. 98, 4032, Hungary
| | - József Virga
- Department of Neurosurgery, University of Debrecen, Clinical Center, Debrecen, Nagyerdei krt. 98, 4032, Hungary
| | - Tibor Hortobágyi
- Department of Neuropathology, University of Debrecen, Clinical Center, Debrecen, Nagyerdei krt. 98, 4032, Hungary
| | - Ádám Dér
- Department of Radiotherapy, University of Debrecen, Clinical Center, Debrecen, Nagyerdei krt. 98, 4032, Hungary
| | - Csaba Szemcsák
- Department of Neurosurgery, University of Debrecen, Clinical Center, Debrecen, Nagyerdei krt. 98, 4032, Hungary
| | - Judit Reményi-Puskár
- Department of Neurosurgery, University of Debrecen, Clinical Center, Debrecen, Nagyerdei krt. 98, 4032, Hungary
| | - László Bognár
- Department of Neurosurgery, University of Debrecen, Clinical Center, Debrecen, Nagyerdei krt. 98, 4032, Hungary
| |
Collapse
|
95
|
Abbassy M, Missios S, Barnett GH, Brewer C, Peereboom DM, Ahluwalia M, Neyman G, Chao ST, Suh JH, Vogelbaum MA. Phase I Trial of Radiosurgery Dose Escalation Plus Bevacizumab in Patients With Recurrent/Progressive Glioblastoma. Neurosurgery 2019; 83:385-392. [PMID: 28973311 DOI: 10.1093/neuros/nyx369] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/25/2017] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The effectiveness of stereotactic radiosurgery (SRS) for recurrent glioblastoma (rGBM) remains uncertain. SRS has been associated with a high risk of radionecrosis in gliomas. OBJECTIVE To determine the safety of dose escalation of single-fraction radiosurgery for rGBM in the setting of bevacizumab therapy. METHODS We conducted a prospective trial to determine the safety and synergistic benefit of higher doses of SRS administered with bevacizumab for rGBM. A single dose of bevacizumab was given prior to SRS and continued until progression. Dose-limiting toxicity was evaluated in successive cohorts of 3 patients. RESULTS Seven males and 2 females entered the study. The maximum linear diameter of the enhancing tumor was 2.58 cm (2.04-3.09). Prescription dose was escalated from 18 to 22 Gy. The radiosurgery target was chosen before the first dose of bevacizumab, about 1 wk prior to SRS treatment. Pre-SRS bevacizumab treatment was associated with a reduction of the mean volume of the enhancing lesion from 4.7 to 2.86 cm3 on the day of SRS (P = .103). No patient developed an acute side effect related to SRS treatment. The combination of SRS and bevacizumab resulted in a partial response in 3 patients and stable disease in 6 patients. Median progression-free and overall survival were 7.5 and 13 mo, respectively. CONCLUSION A single dose of bevacizumab prior to SRS permitted safe prescription dose escalation up to 22 Gy for rGBM. Further evaluation of the efficacy of SRS for rGBM should be performed in the setting of bevacizumab treatment.
Collapse
Affiliation(s)
- Mahmoud Abbassy
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Neurosurgery, Alexandria University, Alexandria, Egypt
| | - Symeon Missios
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| | - Gene H Barnett
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| | - Cathy Brewer
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| | - David M Peereboom
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Manmeet Ahluwalia
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Gennady Neyman
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Samuel T Chao
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - John H Suh
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Michael A Vogelbaum
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Neurosurgery, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
96
|
Lowe S, Bhat KP, Olar A. Current clinical management of patients with glioblastoma. Cancer Rep (Hoboken) 2019; 2:e1216. [PMID: 32721125 DOI: 10.1002/cnr2.1216] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/11/2019] [Accepted: 04/29/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Glioblastoma (GB) is the most aggressive primary brain tumor, historically resistant to treatment, and with overall fatal outcome. RECENT FINDINGS Recently, several molecular subgroups and rare genetic alterations have been described in GB. In this review article, we will describe the current clinical management of patients with GB in the United States, discuss selected next-generation molecular-targeted therapies in GB, and present ongoing clinical trials for patients with GB. This review is intended for clinical and preclinical researchers who conduct work on GB and would like to understand more about the current standard of treatment of GB patients, historical perspectives, current challenges, and ongoing and upcoming clinical trials. CONCLUSIONS GB is an extremely complex disease, and despite recent progress and advanced therapeutic strategies, the overall patient's prognosis remains dismal. Innovative strategies and integrative ways of approach to disease are urgently needed.
Collapse
Affiliation(s)
- Stephen Lowe
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina
| | - Krishna P Bhat
- Deparment of Translational Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adriana Olar
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina.,Departments of Pathology and Laboratory Medicine, Medical University of South Carolina & Hollings Cancer Center, Charleston, South Carolina
| |
Collapse
|
97
|
Wang Y, Pandey M, Ballo MT. Integration of Tumor-Treating Fields into the Multidisciplinary Management of Patients with Solid Malignancies. Oncologist 2019; 24:e1426-e1436. [PMID: 31444292 PMCID: PMC6975944 DOI: 10.1634/theoncologist.2017-0603] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/10/2019] [Indexed: 12/24/2022] Open
Abstract
Tumor treating fields, a noninvasive cancer treatment using low intensity alternating electric fields, offers clinical opportunities with unique challenges. This review focuses on the mechanism of action of this treatment, the known pre‐clinical and clinical experience, and the practical issues surrounding its use in the multidisciplinary management of patients with solid malignancies. Tumor‐treating fields (TTFields) are a noninvasive antimitotic cancer treatment consisting of low‐intensity alternating electric fields delivered to the tumor or tumor bed via externally applied transducer arrays. In multiple in vitro and in vivo cancer cell lines, TTFields therapy inhibits cell proliferation, disrupts cell division, interferes with cell migration and invasion, and reduces DNA repair. Human trials in patients with primary glioblastoma showed an improvement in overall survival, and trials in patients with unresectable malignant pleural mesothelioma showed favorable outcomes compared with historical control. This led to U.S. Food and Drug Administration approval in both clinical situations, paving the way for development of trials investigating TTFields in other malignancies. Although these trials are ongoing, the existing evidence suggests that TTFields have activity outside of neuro‐oncology, and further study into the mechanism of action and clinical activity is required. In addition, because TTFields are a previously unrecognized antimitotic therapy with a unique mode of delivery, the oncological community must address obstacles to widespread patient and provider acceptance. TTFields will likely join surgery, systemic therapy, and radiation therapy as a component of multimodality management of patients with solid malignancies. Implications for Practice. Tumor‐treating fields (TTFields) exhibit a broad range of antitumor activities. Clinically, they improve overall survival for patients with newly diagnosed glioblastoma. The emergence of TTFields has changed the treatment regimen for glioblastoma. Clinicians need to understand the practical issues surrounding its use in the multidisciplinary management of patients with glioblastoma. With ongoing clinical trials, TTFields likely will become another treatment modality for solid malignancies.
Collapse
Affiliation(s)
- Yuefeng Wang
- Department of Radiation Oncology, West Cancer Center and Research Institute, Memphis, Tennessee, USA
| | - Manjari Pandey
- Department of Hematology/Oncology, West Cancer Center and Research Institute, Memphis, Tennessee, USA
| | - Matthew T Ballo
- Department of Radiation Oncology, West Cancer Center and Research Institute, Memphis, Tennessee, USA
| |
Collapse
|
98
|
Geurts M, van den Bent MJ. Timing of radiotherapy in newly diagnosed glioblastoma: no need to rush? Neuro Oncol 2019; 20:868-869. [PMID: 29767783 DOI: 10.1093/neuonc/noy065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Marjolein Geurts
- The Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands.,Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | | |
Collapse
|
99
|
Ludmir EB, Mahajan A, Ahern V, Ajithkumar T, Alapetite C, Bernier-Chastagner V, Bindra RS, Bishop AJ, Bolle S, Brown PD, Carrie C, Chalmers AJ, Chang EL, Chung C, Dieckmann K, Esiashvili N, Gandola L, Ghia AJ, Gondi V, Grosshans DR, Harrabi SB, Horan G, Indelicato DJ, Jalali R, Janssens GO, Krause M, Laack NN, Laperriere N, Laprie A, Li J, Marcus KJ, McGovern SL, Merchant TE, Merrell KW, Padovani L, Parkes J, Paulino AC, Schwarz R, Shih HA, Souhami L, Sulman EP, Taylor RE, Thorp N, Timmermann B, Wheeler G, Wolden SL, Woodhouse KD, Yeboa DN, Yock TI, Kortmann RD, McAleer MF. Assembling the brain trust: the multidisciplinary imperative in neuro-oncology. Nat Rev Clin Oncol 2019; 16:521-522. [PMID: 31150024 DOI: 10.1038/s41571-019-0235-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Ethan B Ludmir
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anita Mahajan
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Verity Ahern
- Sydney West Radiation Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, New South Wales, Australia
| | | | - Claire Alapetite
- Department of Radiation Oncology, Institut Curie, Paris and Orsay, France
| | | | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Andrew J Bishop
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie Bolle
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Christian Carrie
- Department of Radiation Oncology, Centre Léon Bérard, Lyon, France
| | | | - Eric L Chang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Caroline Chung
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karin Dieckmann
- Department of Radiation Oncology, Universität Klinik für Strahlentherapie und Strahlenbiologie, Vienna, Austria
| | - Natia Esiashvili
- Department of Radiation Oncology, Emory University Winship Cancer Institute, Atlanta, GA, USA
| | - Lorenza Gandola
- Pediatric Radiotherapy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Amol J Ghia
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vinai Gondi
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David R Grosshans
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Semi B Harrabi
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Gail Horan
- Department of Oncology, Cambridge University Hospitals, Cambridge, UK
| | - Danny J Indelicato
- Department of Radiation Oncology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Rakesh Jalali
- Department of Radiation Oncology, Apollo Proton Cancer Centre, Chennai, Tamil Nadu, India
| | - Geert O Janssens
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mechthild Krause
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Normand Laperriere
- Department of Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anne Laprie
- Department of Radiation Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Jing Li
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karen J Marcus
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susan L McGovern
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Laetitia Padovani
- Department of Radiation Oncology, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Jeannette Parkes
- Department of Radiation Oncology, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - Arnold C Paulino
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rudolf Schwarz
- Department of Radiation Oncology, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Helen A Shih
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Luis Souhami
- Department of Radiation Oncology, McGill University Health Centre, Cedars Cancer Centre, Montréal, Quebec, Canada
| | - Erik P Sulman
- Department of Radiation Oncology, New York University Langone School of Medicine, New York, NY, USA
| | | | - Nicola Thorp
- Department of Oncology, The Clatterbridge Cancer Centre, Liverpool, UK
| | - Beate Timmermann
- Department of Particle Therapy, University Hospital Essen, West German Proton Therapy Centre Essen, West German Cancer Center, German Cancer Consortium, Essen, Germany
| | - Greg Wheeler
- Department of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kristina D Woodhouse
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Debra N Yeboa
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Torunn I Yock
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Mary Frances McAleer
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
100
|
Ballo MT, Urman N, Lavy-Shahaf G, Grewal J, Bomzon Z, Toms S. Correlation of Tumor Treating Fields Dosimetry to Survival Outcomes in Newly Diagnosed Glioblastoma: A Large-Scale Numerical Simulation-Based Analysis of Data from the Phase 3 EF-14 Randomized Trial. Int J Radiat Oncol Biol Phys 2019; 104:1106-1113. [DOI: 10.1016/j.ijrobp.2019.04.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/16/2019] [Accepted: 04/14/2019] [Indexed: 10/27/2022]
|