701
|
Shevtsov M, Nikolaev B, Marchenko Y, Yakovleva L, Skvortsov N, Mazur A, Tolstoy P, Ryzhov V, Multhoff G. Targeting experimental orthotopic glioblastoma with chitosan-based superparamagnetic iron oxide nanoparticles (CS-DX-SPIONs). Int J Nanomedicine 2018; 13:1471-1482. [PMID: 29559776 PMCID: PMC5856030 DOI: 10.2147/ijn.s152461] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Glioblastoma is the most devastating primary brain tumor of the central nervous system in adults. Magnetic nanocarriers may help not only for a targeted delivery of chemotherapeutic agents into the tumor site but also provide contrast enhancing properties for diagnostics using magnetic resonance imaging (MRI). Methods Synthesized hybrid chitosan-dextran superparamagnetic nanoparticles (CS-DX-SPIONs) were characterized using transmission electron microscopy (TEM) and relaxometry studies. Nonlinear magnetic response measurements were employed for confirming the superparamagnetic state of particles. Following in vitro analysis of nanoparticles cellular uptake tumor targeting was assessed in the model of the orthotopic glioma in rodents. Results CS-DX-SPIONs nanoparticles showed a uniform diameter of 55 nm under TEM and superparamagentic characteristics as determined by T1 (spin-lattice relaxation time) and T2 (spin-spin relaxation time) proton relaxation times. Application of the chitosan increased the charge from +8.9 to +19.3 mV of the dextran-based SPIONs. The nonlinear magnetic response at second harmonic of CS-DX-SPIONs following the slow change of stationary magnetic fields with very low hysteresis evidenced superparamagnetic state of particles at ambient temperatures. Confocal microscopy and flow cytometry studies showed an enhanced internalization of the chitosan-based nanoparticles in U87, C6 glioma and HeLa cells as compared to dextran-coated particles. Cytotoxicity assay demonstrated acceptable toxicity profile of the synthesized nanoparticles up to a concentration of 10 μg/ml. Intravenously administered CS-DX-SPIONs in orthotopic C6 gliomas in rats accumulated in the tumor site as shown by high-resolution MRI (11.0 T). Retention of nanoparticles resulted in a significant contrast enhancement of the tumor image that was accompanied with a dramatic drop in T2 values (P<0.001). Subsequent histological studies proved the accumulation of the nanoparticles inside glioblastoma cells. Conclusion Hybrid chitosan-dextran magnetic particles demonstrated high MR contrast enhancing properties for the delineation of the brain tumor. Due to a significant retention of the particles in the tumor an application of the CS-DX-SPIONs could not only improve the tumor imaging but also could allow a targeted delivery of chemotherapeutic agents.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Department of Cell Biotechnology, Institute of Cytology of the Russian Academy of Sciences, St Petersburg, Russia.,Department of Radiation Immuno Oncology, Technische Universität München, Klinikum rechts der Isar, Munich, Germany.,Department of Biotechnology, Pavlov First Saint Petersburg State Medical University, St Petersburg, Russia.,Department of Pediatric Neurosurgery, Polenov Russian Scientific Research Institute of Neurosurgery, St Petersburg, Russia
| | - Boris Nikolaev
- Department of Nanomedicine, Research Institute of Highly Pure Biopreparations, St Petersburg, Russia
| | - Yaroslav Marchenko
- Department of Nanomedicine, Research Institute of Highly Pure Biopreparations, St Petersburg, Russia
| | - Ludmila Yakovleva
- Department of Nanomedicine, Research Institute of Highly Pure Biopreparations, St Petersburg, Russia
| | - Nikita Skvortsov
- Department of Nanomedicine, Research Institute of Highly Pure Biopreparations, St Petersburg, Russia
| | - Anton Mazur
- Department of NMR, Saint Petersburg State University, St Petersburg, Russia
| | - Peter Tolstoy
- Department of NMR, Saint Petersburg State University, St Petersburg, Russia
| | - Vyacheslav Ryzhov
- Department of NMR, NRC "Kurchatov Institute", Petersburg Nuclear Physics Institute, Gatchina, Russia
| | - Gabriele Multhoff
- Department of Radiation Immuno Oncology, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| |
Collapse
|
702
|
Abstract
Glioblastoma multiforme are mortifying brain tumors that contain a subpopulation of tumor cells with stem-like properties, termed as glioblastoma stem-like cells (GSCs). These GSCs constitute an autonomous reservoir of aberrant cells able to initiate, maintain, and repopulate the tumor mass. A new therapeutic strategy would consist of targeting the GSC population. The GSCs are situated in perivascular niches, closely associated with brain microvascular endothelial cells thereby involved in bidirectional molecular and cellular interactions. In this scenario, the endothelium not only supplies oxygen and necessary nutrients but also seeds a protective microenvironment for tumor growth. Although GSC fate, plasticity, and survival are regulated by external cues emanating from endothelial cells, the nature of such angiocrine signals remains unknown. Our laboratory conclusively demonstrated that brain endothelial cells positively control the expansion of GSCs.1 Notably, we found that GSCs are addicted to the hormonal peptide apelin (APLN) secreted by surrounding endothelial cells, and identified the APLN/APLNR nexus as a promising druggable network in glioblastoma.
Collapse
Affiliation(s)
- Elizabeth Harford-Wright
- CRCINA, Team SOAP, Inserm, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Julie Gavard
- CRCINA, Team SOAP, Inserm, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,Institut de Cancerologie de l'Ouest, Rene Gauducheau, Saint-Herblain, France
| |
Collapse
|
703
|
Youland RS, Lee JY, Kreofsky CR, Brown PD, Uhm JH, Laack NN. Modern reirradiation for recurrent gliomas can safely delay tumor progression. Neurooncol Pract 2018; 5:46-55. [PMID: 31385961 PMCID: PMC6655388 DOI: 10.1093/nop/npx014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Despite advances in modern therapy, high-grade gliomas continue to portend a dismal prognosis and nearly all patients will experience relapse. Unfortunately, salvage options remain limited. In this study, we assessed outcomes for patients with recurrent gliomas treated with reirradiation. METHODS We retrospectively identified 48 glioma patients treated with reirradiation between 2013 and 2016. All had radiographic or pathologic evidence of recurrence. Prognostic factors were abstracted from the electronic medical record. RESULTS Initial surgery included biopsy in 15, subtotal resection in 21, and gross total resection in 12. Initial chemotherapy included temozolomide (TMZ) in 31, TMZ+dasatinib in 7, TMZ+vorinostat in 3, and procarbazine, lomustine, and vincristine in 2. The median dose of primary radiotherapy was 60 Gy delivered in 30 fractions. Median overall survival (OS) and progression-free survival (PFS) from initial diagnosis were 3.2 and 1.7 years, respectively. A total of 36 patients failed salvage bevacizumab before reirradiation. Salvage surgery was performed before reirradiation in 21 patients. Median time to reirradiation was 1.7 years. Median follow-up was 13.7 months from reirradiation. Concurrent systemic therapy was given in 33 patients (bevacizumab in 27, TMZ in 8, and lomustine in 2). Median PFS and OS after reirradiation were 3.2 and 6.3 months, respectively. Radionecrosis occurred in 4 patients and no radionecrosis was seen in patients receiving concurrent bevacizumab with reirradiation (0% vs 19%, P = .03). CONCLUSIONS Reirradiation may result in delayed tumor progression with acceptable toxicity. Prospective trials are needed to determine the impact of reirradiation on tumor progression and quality of life.
Collapse
Affiliation(s)
- Ryan S Youland
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - John Y Lee
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - Cole R Kreofsky
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - Joon H Uhm
- Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| |
Collapse
|
704
|
Straube C, Oechsner M, Kampfer S, Scharl S, Schmidt-Graf F, Wilkens JJ, Combs SE. Dosimetric impact of tumor treating field (TTField) transducer arrays onto treatment plans for glioblastomas - a planning study. Radiat Oncol 2018; 13:31. [PMID: 29471879 PMCID: PMC5824562 DOI: 10.1186/s13014-018-0976-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 02/15/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Tumor-Treating Fields (TTFields) are a novel treatment strategy for glioblastoma (GBM) that is approved for the use concomitantly to adjuvant chemotherapy. Preclinical data suggest a synergistic interaction of TTFields and radiotherapy (RT). However, the dosimetric uncertainties caused by the highly dense arrays have led to caution of applying the TTF setup during RT. METHODS In a RW3 slab phantom we compared the MV- and kV-CT based planned dose with the measured dose. VMAT-plans were optimized on MV-CTs of an Alderson head phantom without TTF arrays and then re-calculated on the same phantom equipped with TTF arrays. Dose at organs at risk (OAR) and target volumes (PTVs) were compared. RESULTS Measurements at a depth of 2, 3 and 4 cm of a RW 3 slab phantom show an attenuation due to TTField arrays of 3.4, 3.7 and 2.7% respectively. This was in-line with calculated attenuations based on MV-CT (1.2, 2.5 and 2.5%) but not with the attenuation expected from kV-CT based calculations (7.1, 8.2 and 8.6%). Consecutive MV-CT based VMAT planning and re-calculation reveals, that the conformity and homogeneity are not affected by the presence of TTField arrays. The dose at organs at risk (OAR) can show increases or decreases by < 0.5 Gy, which should be considered especially in cases next to the scull base. CONCLUSION MV-CT based dose calculation results in reliable dose distributions also in the presence of TTField arrays. There is a small but clinically not relevant interaction between the TTField arrays and VMAT dose application. Thus, daily replacement of TTField arrays is not necessary in regard to deeply located OARs. RT is feasible, when a VMAT treatment plan is optimized to an array free planning CT. As the biologic effect of a concomitant treatment especially on OARs is currently unknown, a concomitant treatment should be performed only within clinical trials.
Collapse
Affiliation(s)
- Christoph Straube
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
| | - Markus Oechsner
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Severin Kampfer
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Sophia Scharl
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Friederike Schmidt-Graf
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Jan J. Wilkens
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Department of Radiation Sciences (DRS), Institute for Innovative Radiotherapy (iRT), Helmholtz Zentrum München, Munich, Germany
| | - Stephanie E. Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, Munich, Germany
- Department of Radiation Sciences (DRS), Institute for Innovative Radiotherapy (iRT), Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
705
|
Radiation Oncology: What's in a Name? Pract Radiat Oncol 2018; 9:125-127. [PMID: 29452871 DOI: 10.1016/j.prro.2017.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/05/2017] [Accepted: 12/11/2017] [Indexed: 11/22/2022]
|
706
|
β-escin selectively targets the glioblastoma-initiating cell population and reduces cell viability. Oncotarget 2018; 7:66865-66879. [PMID: 27589691 PMCID: PMC5341843 DOI: 10.18632/oncotarget.11784] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/10/2016] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive tumour of the central nervous system and is associated with an extremely poor prognosis. Within GBM exists a subpopulation of cells, glioblastoma-initiating cells (GIC), which possess the characteristics of progenitor cells, have the ability to initiate tumour growth and resist to current treatment strategies. We aimed at identifying novel specific inhibitors of GIC expansion through use of a large-scale chemical screen of approved small molecules. Here, we report the identification of the natural compound β-escin as a selective inhibitor of GIC viability. Indeed, β-escin was significantly cytotoxic in nine patient-derived GIC, whilst exhibiting no substantial effect on the other human cancer or control cell lines tested. In addition, β-escin was more effective at reducing GIC growth than current clinically used cytotoxic agents. We further show that β-escin triggers caspase-dependent cell death combined with a loss of stemness properties. However, blocking apoptosis could not rescue the β-escin-induced reduction in sphere formation or stemness marker activity, indicating that β-escin directly modifies the stem identity of GIC, independent of the induction of cell death. Thus, this study has repositioned β-escin as a promising potential candidate to selectively target the aggressive population of initiating cells within GBM.
Collapse
|
707
|
Brown PD, Ahluwalia MS, Khan OH, Asher AL, Wefel JS, Gondi V. Whole-Brain Radiotherapy for Brain Metastases: Evolution or Revolution? J Clin Oncol 2018; 36:483-491. [PMID: 29272161 PMCID: PMC6075843 DOI: 10.1200/jco.2017.75.9589] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
An estimated 20% of patients with cancer will develop brain metastases. Approximately 200,000 individuals in the United States alone receive whole-brain radiotherapy (WBRT) each year to treat brain metastases. Historically, the prognosis of patients with brain metastases has been poor; however, with new therapies, this is changing. Because patients are living longer following the diagnosis and treatment of brain metastases, there has been rising concern about treatment-related toxicities associated with WBRT, including neurocognitive toxicity. In addition, recent clinical trials have raised questions about the use of WBRT. To better understand this rapidly changing landscape, this review outlines the treatment roles and toxicities of WBRT and alternative therapies for the management of brain metastases.
Collapse
Affiliation(s)
- Paul D. Brown
- Paul D. Brown, Mayo Clinic, Rochester, MN; Manmeet S. Ahluwalia, Cleveland Clinic, Cleveland, OH; Osaama H. Khan and Vinai Gondi, Northwestern University Feinberg School of Medicine, Chicago; Vinai Gondi, Northwestern Medicine Cancer Center, Warrenville, IL; Anthony L. Asher, Carolina Neurosurgery and Spine Associates and Neurosciences Institute, Carolinas HealthCare System, Charlotte, NC; and Jeffrey S. Wefel, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Manmeet S. Ahluwalia
- Paul D. Brown, Mayo Clinic, Rochester, MN; Manmeet S. Ahluwalia, Cleveland Clinic, Cleveland, OH; Osaama H. Khan and Vinai Gondi, Northwestern University Feinberg School of Medicine, Chicago; Vinai Gondi, Northwestern Medicine Cancer Center, Warrenville, IL; Anthony L. Asher, Carolina Neurosurgery and Spine Associates and Neurosciences Institute, Carolinas HealthCare System, Charlotte, NC; and Jeffrey S. Wefel, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Osaama H. Khan
- Paul D. Brown, Mayo Clinic, Rochester, MN; Manmeet S. Ahluwalia, Cleveland Clinic, Cleveland, OH; Osaama H. Khan and Vinai Gondi, Northwestern University Feinberg School of Medicine, Chicago; Vinai Gondi, Northwestern Medicine Cancer Center, Warrenville, IL; Anthony L. Asher, Carolina Neurosurgery and Spine Associates and Neurosciences Institute, Carolinas HealthCare System, Charlotte, NC; and Jeffrey S. Wefel, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anthony L. Asher
- Paul D. Brown, Mayo Clinic, Rochester, MN; Manmeet S. Ahluwalia, Cleveland Clinic, Cleveland, OH; Osaama H. Khan and Vinai Gondi, Northwestern University Feinberg School of Medicine, Chicago; Vinai Gondi, Northwestern Medicine Cancer Center, Warrenville, IL; Anthony L. Asher, Carolina Neurosurgery and Spine Associates and Neurosciences Institute, Carolinas HealthCare System, Charlotte, NC; and Jeffrey S. Wefel, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeffrey S. Wefel
- Paul D. Brown, Mayo Clinic, Rochester, MN; Manmeet S. Ahluwalia, Cleveland Clinic, Cleveland, OH; Osaama H. Khan and Vinai Gondi, Northwestern University Feinberg School of Medicine, Chicago; Vinai Gondi, Northwestern Medicine Cancer Center, Warrenville, IL; Anthony L. Asher, Carolina Neurosurgery and Spine Associates and Neurosciences Institute, Carolinas HealthCare System, Charlotte, NC; and Jeffrey S. Wefel, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vinai Gondi
- Paul D. Brown, Mayo Clinic, Rochester, MN; Manmeet S. Ahluwalia, Cleveland Clinic, Cleveland, OH; Osaama H. Khan and Vinai Gondi, Northwestern University Feinberg School of Medicine, Chicago; Vinai Gondi, Northwestern Medicine Cancer Center, Warrenville, IL; Anthony L. Asher, Carolina Neurosurgery and Spine Associates and Neurosciences Institute, Carolinas HealthCare System, Charlotte, NC; and Jeffrey S. Wefel, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
708
|
MicroRNA-Based Drugs for Brain Tumors. Trends Cancer 2018; 4:222-238. [PMID: 29506672 DOI: 10.1016/j.trecan.2017.12.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 12/18/2017] [Accepted: 12/24/2017] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are key regulatory elements encoded by the genome. A single miRNA can downregulate the expression of multiple genes involved in diverse functions. Because cancer is a disease with multiple gene aberrations, developing novel approaches to identify and modulate miRNA pathways may result in a breakthrough for cancer treatment. With a special focus on glioblastoma (GBM), this review provides an up-to-date summary of miRNA biogenesis, the role of miRNA in cancer resistance, and essential tools for modulating miRNA expression, as well as of clinically promising RNAi delivery systems and how they can be adapted for therapy.
Collapse
|
709
|
Abstract
Glioblastoma is the most common and most aggressive form of primary brain tumor in adults and contributes to high social and medical burden as a result of its incurable nature and significant neurologic morbidity. Despite ongoing research, there has not been improvement in survival in glioblastoma. This review discusses recent advances in clinically significant molecular profiling, including IDH mutation status and O6-methylguanine-DNA methyltransferase ( MGMT) promoter methylation. We review updates in management of newly diagnosed and recurrent glioblastoma, as well as common difficulties in management, such as pseudoprogression and pseudoresponse. Ongoing translational research in targeted therapy and immunotherapy is briefly discussed.
Collapse
Affiliation(s)
- Joo Yeon Nam
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - John F de Groot
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
710
|
Skolnik AD, Wang S, Gopal PP, Mohan S. Commentary: Pitfalls in the Neuroimaging of Glioblastoma in the Era of Antiangiogenic and Immuno/Targeted Therapy. Front Neurol 2018; 9:51. [PMID: 29459848 PMCID: PMC5807681 DOI: 10.3389/fneur.2018.00051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 01/18/2018] [Indexed: 12/22/2022] Open
Affiliation(s)
- Aaron D Skolnik
- Radiology, Penn Medicine Princeton Health, Plainsboro, NJ, United States
| | - Sumei Wang
- Neuroradiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Pallavi P Gopal
- Pathology, Yale School of Medicine, New Haven, CT, United States
| | - Suyash Mohan
- Neuroradiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
711
|
Burri SH, Gondi V, Brown PD, Mehta MP. The Evolving Role of Tumor Treating Fields in Managing Glioblastoma: Guide for Oncologists. Am J Clin Oncol 2018; 41:191-196. [PMID: 28832384 PMCID: PMC5779316 DOI: 10.1097/coc.0000000000000395] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Glioblastoma (GBM) is a devastating brain tumor with poor prognosis despite advances in surgery, radiation, and chemotherapy. Survival of patients with glioblastoma remains poor, with only 1 in 4 patients alive at 2 years, and a 5-year survival rate of about 5%. Recurrence is nearly universal and, after recurrence, prognosis is poor with very short progression-free survival and overall survival (OS). Various salvage chemotherapy strategies have been applied with limited success. Tumor Treating Fields (TTFields) are a novel treatment modality approved for treatment of either newly diagnosed or recurrent GBM. TTFields therapy involves a medical device and transducer arrays to provide targeted delivery of low intensity, intermediate frequency, alternating electric fields to produce antimitotic effects selective for rapidly dividing tumor cells with limited toxicity. In the phase 3 EF-14 trial, TTFields plus temozolomide provided significantly longer progression-free survival and OS compared with temozolomide alone in patients with newly diagnosed GBM after initial chemoradiotherapy. The addition of TTFields to standard therapy improved median OS from 15.6 to 20.5 months (P=0.04). In the phase 3 EF-11 trial, for recurrent GBM, TTFields provided comparable efficacy as investigator's choice systemic therapy, with improved patient-reported quality of life and a lower incidence of serious adverse events. Primary toxicity associated with TTFields is skin irritation generally managed with array relocation and topical treatments including antibiotics and steroids. TTFields therapy has demonstrated proven efficacy in management of GBM, including improvement in OS for patients with newly diagnosed GBM, and is under current investigation in other brain and extracranial tumors.
Collapse
Affiliation(s)
- Stuart H. Burri
- Levine Cancer Institute
- Southeast Radiation Oncology Group, Charlotte, NC
| | - Vinai Gondi
- Brain & Spine Tumor Center, Northwestern Medicine Cancer Center Warrenville
- Northwestern Medicine Chicago Proton Center, Chicago, IL
| | - Paul D. Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - Minesh P. Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Miami, FL
| |
Collapse
|
712
|
Effect of Radiation Treatment Volume Reduction on Lymphopenia in Patients Receiving Chemoradiotherapy for Glioblastoma. Int J Radiat Oncol Biol Phys 2018; 101:217-225. [PMID: 29502931 DOI: 10.1016/j.ijrobp.2018.01.069] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/31/2017] [Accepted: 01/22/2018] [Indexed: 11/21/2022]
Abstract
PURPOSE To evaluate whether reduction in glioblastoma radiation treatment volume can reduce risk of acute severe lymphopenia (ASL). METHODS AND MATERIALS A total of 210 patients with supratentorial/nonmetastatic glioblastoma were treated with radiation therapy (RT) plus temozolomide from 2007 to 2016 and had laboratory data on total lymphocyte counts. Before 2015, 164 patients were treated with standard-field RT (SFRT), and limited-field RT (LFRT) was implemented thereafter for 46 patients to reduce treatment volume. Total lymphocyte counts were evaluated at baseline, during RT, and at approximately week 12 from initiating RT. Acute severe lymphopenia was defined as any total lymphocyte count < 500 cells/μL within 3 months (by week 12) of initiating RT. Multivariate analysis for overall survival (OS) was performed with Cox regression and with logistic regression for ASL. Propensity score matching was performed to adjust for variability between cohorts. Acute severe lymphopenia, progression-free survival (PFS), and OS were compared using the Kaplan-Meier method. RESULTS Limited-field RT patients had higher gross tumor volume than SFRT patients yet lower brain dose-volume parameters, including volume receiving 25 Gy (V25 Gy: 41% vs 53%, respectively, P < .01). Total lymphocyte count at week 12 was significantly higher for LFRT than for SFRT (median: 1100 cells/μL vs 900 cells/μL, respectively, P = .02). On multivariate analysis, ASL was an independent predictor of OS, and brain V25 Gy was an independent predictor of ASL. The ASL rate at 3 months was 15.5% for LFRT and 33.8% for SFRT (P = .12). In a propensity-matched comparison of 45 pairs of LFRT and SFRT patients, PFS (median: 5.9 vs 6.2 months, respectively, P = .58) and OS (median: 16.2 vs 13.9 months, respectively, P = .69) were not significantly different. CONCLUSIONS Limited-field RT is associated with less lymphopenia after RT plus temozolomide and does not adversely affect PFS or OS. Brain V25 Gy is confirmed as an important dosimetric predictor for ASL.
Collapse
|
713
|
Jackson WC, Tsien CI, Junck L, Leung D, Hervey-Jumper S, Orringer D, Heth J, Wahl DR, Spratt DE, Cao Y, Lawrence TS, Kim MM. Standard dose and dose-escalated radiation therapy are associated with favorable survival in select elderly patients with newly diagnosed glioblastoma. J Neurooncol 2018; 138:155-162. [DOI: 10.1007/s11060-018-2782-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/29/2018] [Indexed: 11/24/2022]
|
714
|
Blumenthal DT, Kanner AA, Aizenstein O, Cagnano E, Greenberg A, Hershkovitz D, Ram Z, Bokstein F. Surgery for Recurrent High-Grade Glioma After Treatment with Bevacizumab. World Neurosurg 2018; 110:e727-e737. [DOI: 10.1016/j.wneu.2017.11.105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/16/2017] [Accepted: 11/18/2017] [Indexed: 01/04/2023]
|
715
|
Qazi MA, Vora P, Venugopal C, Sidhu SS, Moffat J, Swanton C, Singh SK. Intratumoral heterogeneity: pathways to treatment resistance and relapse in human glioblastoma. Ann Oncol 2018; 28:1448-1456. [PMID: 28407030 DOI: 10.1093/annonc/mdx169] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Indexed: 01/01/2023] Open
Abstract
Intratumoral heterogeneity (ITH) has increasingly being described for multiple cancers as the root cause of therapy resistance. Recent studies have started to explore the scope of ITH in glioblastoma (GBM), a highly aggressive and fatal form of brain tumor, to explain its inevitable therapy resistance and disease relapse. In this review, we detail the emerging data that explores the extensive genetic, cellular and functional ITH present in GBM. We discuss current experimental models of human GBM recurrence and suggest harnessing new technologies (CRISPR-Cas9 screening, CyTOF, cellular barcoding, single cell analysis) to delineate GBM ITH and identify treatment-refractory cell populations, thus opening new therapeutic windows. We will also explore why current therapeutics have failed in clinical trials and how ITH can inform us on developing empiric therapies for the treatment of recurrent GBM.
Collapse
Affiliation(s)
- M A Qazi
- Stem Cell and Cancer Research Institute.,Department of Biochemistry and Biomedical Sciences
| | - P Vora
- Stem Cell and Cancer Research Institute.,Department of Surgery, McMaster University, Hamilton
| | - C Venugopal
- Stem Cell and Cancer Research Institute.,Department of Surgery, McMaster University, Hamilton
| | - S S Sidhu
- Donnelly Centre and Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - J Moffat
- Donnelly Centre and Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - C Swanton
- The Francis Crick Institute, University College London Institute, London, UK
| | - S K Singh
- Stem Cell and Cancer Research Institute.,Department of Biochemistry and Biomedical Sciences.,Department of Surgery, McMaster University, Hamilton
| |
Collapse
|
716
|
Dolera M, Malfassi L, Carrara N, Finesso S, Marcarini S, Mazza G, Pavesi S, Sala M, Urso G. Volumetric Modulated Arc (Radio) Therapy in Pets Treatment: The "La Cittadina Fondazione" Experience. Cancers (Basel) 2018; 10:E30. [PMID: 29364837 PMCID: PMC5836062 DOI: 10.3390/cancers10020030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/29/2017] [Accepted: 01/16/2018] [Indexed: 12/31/2022] Open
Abstract
Volumetric Modulated Arc Therapy (VMAT) is a modern technique, widely used in human radiotherapy, which allows a high dose to be delivered to tumor volumes and low doses to the surrounding organs at risk (OAR). Veterinary clinics takes advantage of this feature due to the small target volumes and distances between the target and the OAR. Sparing the OAR permits dose escalation, and hypofractionation regimens reduce the number of treatment sessions with a simpler manageability in the veterinary field. Multimodal volumes definition is mandatory for the small volumes involved and a positioning device precisely reproducible with a setup confirmation is needed before each session for avoiding missing the target. Additionally, the elaborate treatment plan must pursue hard constraints and objectives, and its feasibility must be evaluated with a per patient quality control. The aim of this work is to report results with regard to brain meningiomas and gliomas, trigeminal nerve tumors, brachial plexus tumors, adrenal tumors with vascular invasion and rabbit thymomas, in comparison with literature to determine if VMAT is a safe and viable alternative to surgery or chemotherapy alone, or as an adjuvant therapy in pets.
Collapse
Affiliation(s)
- Mario Dolera
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Luca Malfassi
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Nancy Carrara
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Sara Finesso
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Silvia Marcarini
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Giovanni Mazza
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Simone Pavesi
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Massimo Sala
- La Cittadina Fondazione Studi e Ricerche Veterinarie, 26014 Romanengo, Italy.
| | - Gaetano Urso
- Azienda Socio Sanitaria Territoriale della provincia di Lodi, 26841 Casalpusterlengo, Italy.
| |
Collapse
|
717
|
Wick A, Kessler T, Elia AEH, Winkler F, Batchelor TT, Platten M, Wick W. Glioblastoma in elderly patients: solid conclusions built on shifting sand? Neuro Oncol 2018; 20:174-183. [PMID: 29016815 PMCID: PMC5777484 DOI: 10.1093/neuonc/nox133] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Management of glioblastoma in the elderly population is challenging. In the near future, more than half of patients with this tumor will be over the age of 65. Clinicians have been historically reluctant to treat such patients with the same intensity as younger patients. Due to upper age limits or poor accrual of elderly patients in clinical trials, randomized data for this patient population have been relatively sparse until recently. In this review, we will discuss the concept of an elderly patient population, describe evidence for molecular differences in glioblastoma of elderly versus young patients, evaluate recent first-line trials studying glioblastoma in elderly patients, and discuss best therapeutic practices including the value of molecular testing.
Collapse
Affiliation(s)
- Antje Wick
- Neurology Clinic & German Consortium for Translational Cancer Research (DKTK), Heidelberg University Medical Center & DKFZ, Heidelberg, Germany
| | - Tobias Kessler
- Neurology Clinic & German Consortium for Translational Cancer Research (DKTK), Heidelberg University Medical Center & DKFZ, Heidelberg, Germany
| | - Andrew E H Elia
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Frank Winkler
- Neurology Clinic & German Consortium for Translational Cancer Research (DKTK), Heidelberg University Medical Center & DKFZ, Heidelberg, Germany
| | - Tracy T Batchelor
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Wolfgang Wick
- Neurology Clinic & German Consortium for Translational Cancer Research (DKTK), Heidelberg University Medical Center & DKFZ, Heidelberg, Germany
| |
Collapse
|
718
|
Combination Therapy with Sulfasalazine and Valproic Acid Promotes Human Glioblastoma Cell Death Through Imbalance of the Intracellular Oxidative Response. Mol Neurobiol 2018; 55:6816-6833. [PMID: 29349577 DOI: 10.1007/s12035-018-0895-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 01/09/2018] [Indexed: 01/15/2023]
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor and still lacks effective therapeutic strategies. It has already been shown that old drugs like sulfasalazine (SAS) and valproic acid (VPA) present antitumoral activities in glioma cell lines. SAS has also been associated with a decrease of intracellular glutathione (GSH) levels through a potent inhibition of xc- glutamate/cystine exchanger leading to an antioxidant deprotection. In the same way, VPA was recently identified as a histone deacetylase (HDAT) inhibitor capable of activating tumor suppression genes. As both drugs are widely used in clinical practice and their profile of adverse effects is well known, the aim of our study was to investigate the effects of the combined treatment with SAS and VPA in GBM cell lines. We observed that both drugs were able to reduce cell viability in a dose-dependent manner and the combined treatment potentiated these effects. Combined treatment also increased cell death and inhibited proliferation of GBM cells, while having no effect on human and rat cultured astrocytes. Also, we observed high protein expression of the catalytic subunit of xc- in all the examined GBM cell lines, and treatment with SAS blocked its activity and decreased intracellular GSH levels. Noteworthy, SAS but not VPA was also able to reduce the [14C]-ascorbate uptake. Together, these data indicate that SAS and VPA exhibit a substantial effect on GBM cell's death related to an intracellular oxidative response imbalance, making this combination of drugs a promising therapeutic strategy.
Collapse
|
719
|
Tumor treating fields: a new approach to glioblastoma therapy. J Neurooncol 2018; 137:447-453. [DOI: 10.1007/s11060-018-2768-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 12/20/2022]
|
720
|
Zygogianni A, Protopapa M, Kougioumtzopoulou A, Simopoulou F, Nikoloudi S, Kouloulias V. From imaging to biology of glioblastoma: new clinical oncology perspectives to the problem of local recurrence. Clin Transl Oncol 2018; 20:989-1003. [PMID: 29335830 DOI: 10.1007/s12094-018-1831-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 01/04/2018] [Indexed: 12/13/2022]
Abstract
GBM is one of the most common and aggressive brain tumors. Surgery and adjuvant chemoradiation have succeeded in providing a survival benefit. Although most patients will eventually experience local recurrence, the means to fight recurrence are limited and prognosis remains poor. In a disease where local control remains the major challenge, few trials have addressed the efficacy of local treatments, either surgery or radiation therapy. The present article reviews recent advances in the biology, imaging and biomarker science of GBM as well as the current treatment status of GBM, providing new perspectives to the problem of local recurrence.
Collapse
Affiliation(s)
- A Zygogianni
- Radiotherapy Unit, 1st Department of Radiology, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - M Protopapa
- Radiotherapy Unit, 1st Department of Radiology, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - A Kougioumtzopoulou
- Radiotherapy Unit, 2nd Department of Radiology, Medical School, ATTIKON University Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462, Chaidari, Greece
| | - F Simopoulou
- Radiotherapy Unit, 1st Department of Radiology, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - S Nikoloudi
- Radiotherapy Unit, 1st Department of Radiology, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - V Kouloulias
- Radiotherapy Unit, 2nd Department of Radiology, Medical School, ATTIKON University Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462, Chaidari, Greece.
| |
Collapse
|
721
|
Youland RS, Pafundi DH, Brinkmann DH, Lowe VJ, Morris JM, Kemp BJ, Hunt CH, Giannini C, Parney IF, Laack NN. Prospective trial evaluating the sensitivity and specificity of 3,4-dihydroxy-6-[18F]-fluoro-L-phenylalanine (18F-DOPA) PET and MRI in patients with recurrent gliomas. J Neurooncol 2018; 137:583-591. [PMID: 29330751 DOI: 10.1007/s11060-018-2750-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 01/07/2018] [Indexed: 12/11/2022]
Abstract
Treatment-related changes can be difficult to differentiate from progressive glioma using MRI with contrast (CE). The purpose of this study is to compare the sensitivity and specificity of 18F-DOPA-PET and MRI in patients with recurrent glioma. Thirteen patients with MRI findings suspicious for recurrent glioma were prospectively enrolled and underwent 18F-DOPA-PET and MRI for neurosurgical planning. Stereotactic biopsies were obtained from regions of concordant and discordant PET and MRI CE, all within regions of T2/FLAIR signal hyperintensity. The sensitivity and specificity of 18F-DOPA-PET and CE were calculated based on histopathologic analysis. Receiver operating characteristic curve analysis revealed optimal tumor to normal (T/N) and SUVmax thresholds. In the 37 specimens obtained, 51% exhibited MRI contrast enhancement (M+) and 78% demonstrated 18F-DOPA-PET avidity (P+). Imaging characteristics included M-P- in 16%, M-P+ in 32%, M+P+ in 46% and M+P- in 5%. Histopathologic review of biopsies revealed grade II components in 16%, grade III in 43%, grade IV in 30% and no tumor in 11%. MRI CE sensitivity for recurrent tumor was 52% and specificity was 50%. PET sensitivity for tumor was 82% and specificity was 50%. A T/N threshold > 2.0 altered sensitivity to 76% and specificity to 100% and SUVmax > 1.36 improved sensitivity and specificity to 94 and 75%, respectively. 18F-DOPA-PET can provide increased sensitivity and specificity compared with MRI CE for visualizing the spatial distribution of recurrent gliomas. Future studies will incorporate 18F-DOPA-PET into re-irradiation target volume delineation for RT planning.
Collapse
Affiliation(s)
- Ryan S Youland
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Deanna H Pafundi
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Debra H Brinkmann
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Val J Lowe
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jonathan M Morris
- Division of Neuroradiology, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Bradley J Kemp
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Christopher H Hunt
- Division of Neuroradiology, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Caterina Giannini
- Department of Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
722
|
Kong XT, Nguyen NT, Choi YJ, Zhang G, Nguyen HN, Filka E, Green S, Yong WH, Liau LM, Green RM, Kaprealian T, Pope WB, Nghiemphu PL, Cloughesy T, Lassman A, Lai A. Phase 2 Study of Bortezomib Combined With Temozolomide and Regional Radiation Therapy for Upfront Treatment of Patients With Newly Diagnosed Glioblastoma Multiforme: Safety and Efficacy Assessment. Int J Radiat Oncol Biol Phys 2018; 100:1195-1203. [PMID: 29722661 DOI: 10.1016/j.ijrobp.2018.01.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/08/2017] [Accepted: 01/02/2018] [Indexed: 11/17/2022]
Abstract
PURPOSE To assess the safety and efficacy of upfront treatment using bortezomib combined with standard radiation therapy (RT) and temozolomide (TMZ), followed by adjuvant bortezomib and TMZ for ≤24 cycles, in patients with newly diagnosed glioblastoma multiforme (GBM). METHODS AND MATERIALS Twenty-four patients with newly diagnosed GBM were enrolled. The patients received standard external beam regional RT with concurrent TMZ beginning 3 to 6 weeks after surgery, followed by adjuvant TMZ and bortezomib for ≤24 cycles or until tumor progression. During RT, bortezomib was given at 1.3 mg/m2 on days 1, 4, 8, 11, 29, 32, 36, and 39. After RT, bortezomib was given at 1.3 mg/m2 on days 1, 4, 8, and 11 every 4 weeks. RESULTS No unexpected adverse events occurred from the addition of bortezomib. The efficacy analysis showed a median progression-free survival (PFS) of 6.2 months (95% confidence interval [CI] 3.7-8.8), with promising PFS rates at ≥18 months compared with historical norms (25.0% at 18 and 24 months; 16.7% at 30 months). In terms of overall survival (OS), the median OS was 19.1 months (95% CI 6.7-31.4), with improved OS rates at ≥12 months (87.5% at 12, 50.0% at 24, 34.1% at 36-60 months) compared with the historical norms. The median PFS was 24.7 months (95% CI 8.5-41.0) in 10 MGMT methylated and 5.1 months (95% CI 3.9-6.2) in 13 unmethylated patients. The estimated median OS was 61 months (95% CI upper bound not reached) in the methylated and 16.4 months (95% CI 11.8-21.0) in the unmethylated patients. CONCLUSIONS The addition of bortezomib to current standard radiochemotherapy in newly diagnosed GBM patients was tolerable. The PFS and OS rates appeared promising, with more benefit to MGMT methylated patients. Further clinical investigation is warranted in a larger cohort of patients.
Collapse
Affiliation(s)
- Xiao-Tang Kong
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California; Department of Neurology, School of Medicine at University of California, Irvine, Irvine, California
| | - Nhung T Nguyen
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Yoon J Choi
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Guicheng Zhang
- School of Public Health, Xinxiang Medical University, Xinxiang, China, and Curtin University, Perth, Western Australia, Australia
| | - HuyTram N Nguyen
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Emese Filka
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Stacey Green
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - William H Yong
- Department of Pathology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Richard M Green
- Kaiser Permanente Southern California, Los Angeles, California
| | - Tania Kaprealian
- Department of Radiation Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Whitney B Pope
- Department of Radiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - P Leia Nghiemphu
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Timothy Cloughesy
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Andrew Lassman
- Department of Neurology, Columbia University, New York, New York
| | - Albert Lai
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
723
|
Dai S, Yan Y, Xu Z, Zeng S, Qian L, Huo L, Li X, Sun L, Gong Z. SCD1 Confers Temozolomide Resistance to Human Glioma Cells via the Akt/GSK3β/β-Catenin Signaling Axis. Front Pharmacol 2018; 8:960. [PMID: 29354058 PMCID: PMC5758607 DOI: 10.3389/fphar.2017.00960] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/18/2017] [Indexed: 02/05/2023] Open
Abstract
Resistance to temozolomide (TMZ), the standard chemotherapy agent for glioblastoma (GBM), poses a major clinical challenge to GBM prognosis. Understanding the mechanisms of TMZ resistance can help to identify novel drug targets and more effective therapies. Recent studies suggest that bioenergetic alterations of cancer cells play important roles in drug resistance. In our study, the altered metabolism of cancer cells was observed using a metabolic PCR array. We found that stearoyl-coenzyme A desaturase 1 (SCD1), a key rate-limiting enzyme for synthesis of monounsaturated fatty acids, was significantly upregulated in TMZ-resistant GBM cells compared to their parental counterparts. Overexpression of SCD1 promoted resistance to TMZ in parental GBM cells, whereas SCD1 downregulation by siRNA could re-sensitize TMZ-resistant cells in vitro. Combinational treatment of TMZ and an SCD1-specific inhibitor showed a combined inhibitory effect on TMZ-resistant glioma cells. We also observed that overexpression of SCD1 promoted Akt/GSK3β/β-catenin signaling, while silencing of SCD1 inhibited the signaling. The combination of an Akt activator with exogenous SCD1 or the combined inhibition of Akt and enforced expression of SCD1 resulted in the most significant changes of Akt signaling. Functionally, significantly lower viability and mobility rates were observed in TMZ-resistant cells when treated with Akt inhibitors and an SCD1 inhibitor simultaneously compared to when treated individually. In conclusion, our study identified SCD1 along with its functional pathway as a novel target in the development of TMZ resistance. SCD1 inhibition used alone or in combination with Akt inhibition could effectively overcome TMZ resistance in gliomas.
Collapse
Affiliation(s)
- Shuang Dai
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- School of Pharmaceutical Sciences, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Long Qian
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Huo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lunquan Sun
- Center for Molecular Medicine, Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
724
|
Meijer DKF, Geesink HJH. Favourable and Unfavourable EMF Frequency Patterns in Cancer: Perspectives for Improved Therapy and Prevention. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/jct.2018.93019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
725
|
Lukas RV, Wu J, Dey M, Buerki RA, Byrne RW, Dohrmann GJ. A Survey of the Neuro-Oncology Landscape. J Clin Neurol 2018; 14:8-15. [PMID: 29141278 PMCID: PMC5765260 DOI: 10.3988/jcn.2018.14.1.8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 11/17/2022] Open
Abstract
The field of neuro-oncology is evolving rapidly. Many important advances have recently been reported, and other promising investigations have the potential to soon make substantial impacts in the field, especially in the areas of high-grade gliomas and brain metastases. We present an overview of the current status of this field, highlighting the key recent advances as well as representative work of key clinical investigations, since these concepts have the potential to influence clinical management if they are demonstrated to be safe and efficacious. This overview includes some work that has only appeared in abstract form in order to provide a timely understanding of how the field is actively changing and what may lie on the horizon. We focus on both medical and surgical neuro-oncology advances in this highly multidisciplinary subspecialty.
Collapse
Affiliation(s)
- Rimas V Lukas
- Department of Neurology, Northwestern University, Chicago, IL, USA.
| | - Jing Wu
- Neuro-Oncology Branch, National Institutes of Health, Bethesda, MD, USA
| | - Mahua Dey
- Department of Neurosurgery, Indiana University, Indianapolis, IN, USA
| | - Robin A Buerki
- Department of Neurosurgery, University of California, San Francisco, CA, USA
| | - Richard W Byrne
- Department of Neurosurgery, Rush University, Chicago, IL, USA
| | | |
Collapse
|
726
|
Wenger C, Miranda PC, Salvador R, Thielscher A, Bomzon Z, Giladi M, Mrugala MM, Korshoej AR. A Review on Tumor-Treating Fields (TTFields): Clinical Implications Inferred From Computational Modeling. IEEE Rev Biomed Eng 2018; 11:195-207. [DOI: 10.1109/rbme.2017.2765282] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
727
|
Sim HW, Morgan ER, Mason WP. Contemporary management of high-grade gliomas. CNS Oncol 2018; 7:51-65. [PMID: 29241354 PMCID: PMC6001673 DOI: 10.2217/cns-2017-0026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/02/2017] [Indexed: 01/01/2023] Open
Abstract
High-grade gliomas, including glioblastoma, are the most common malignant brain tumors in adults. Despite intensive efforts to develop new therapies for these diseases, treatment options remain limited and prognosis is poor. Recently, there have been important advances in our understanding of the molecular basis of glioma, leading to refinements in our diagnostic and management approach. There is new evidence to guide the treatment of elderly patients. A multitude of new agents have been investigated, including targeted therapies, immunotherapeutics and tumor-treating fields. This review summarizes the key findings from this research, and presents a perspective on future opportunities to advance the field.
Collapse
Affiliation(s)
- Hao-Wen Sim
- Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Erin R Morgan
- Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Warren P Mason
- Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| |
Collapse
|
728
|
Schiff D, Schrag D. Living in a material world: tumor-treating fields at the top of the charts. Neuro Oncol 2018; 18:1033-4. [PMID: 27382117 DOI: 10.1093/neuonc/now138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- David Schiff
- University of Virginia Neuro-Oncology Center, Charlottesville, Virginia (D. Schiff); Dana-Farber Cancer Institute, Boston, Massachusetts (D. Schrag)
| | - Deborah Schrag
- University of Virginia Neuro-Oncology Center, Charlottesville, Virginia (D. Schiff); Dana-Farber Cancer Institute, Boston, Massachusetts (D. Schrag)
| |
Collapse
|
729
|
Martínez-Garcia M, Álvarez-Linera J, Carrato C, Ley L, Luque R, Maldonado X, Martínez-Aguillo M, Navarro LM, Vaz-Salgado MA, Gil-Gil M. SEOM clinical guidelines for diagnosis and treatment of glioblastoma (2017). Clin Transl Oncol 2018; 20:22-28. [PMID: 29086250 PMCID: PMC5785619 DOI: 10.1007/s12094-017-1763-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 11/08/2022]
Abstract
Glioblastoma (GB) is the most common brain malignancy and accounts for over 50% of all high-grade gliomas. Radiotherapy (RT) with concomitant and adjuvant temozolomide (TMZ) chemotherapy is the current standard of care for patients with newly diagnosed GB up to age 70. Recently, a new standard of care has been adopted for elderly patients (≥ 65 years) based on short course of RT and TMZ. Several clinically relevant molecular markers that assist in diagnosis and prognosis have recently been identified. The treatment for recurrent GB is not well defined, and decision-making is usually based on prior strategies as well as several clinical and radiological factors. The presence of neurologic deficits and seizures can significantly impact quality of life.
Collapse
Affiliation(s)
| | | | - C. Carrato
- Anatomía Patológica, Hospital Universitari Germans Trias i Pujol de Badalona, Barcelona, Spain
| | - L. Ley
- Neurocirugía, Hospital Ramón y Cajal, Madrid, Spain
| | - R. Luque
- Oncología Médica, Complejo Hospitalario Universitario de Granada Virgen de las Nieves, Granada, Spain
| | - X. Maldonado
- Oncología Radioterápica, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | | | - L. M. Navarro
- Oncología Médica, Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | | | - M. Gil-Gil
- Unidad de Neuro-oncologia. Oncología Médica Institut Català d’Oncologia (ICO)-Hospital Universitari de Bellvitge IDIBELL L’Hospitalet, C/de la Feixa Llarga, s/n, Hospitalet de Llobregat, 08907 Barcelona, Spain
| |
Collapse
|
730
|
Giladi M, Munster M, Schneiderman RS, Voloshin T, Porat Y, Blat R, Zielinska-Chomej K, Hååg P, Bomzon Z, Kirson ED, Weinberg U, Viktorsson K, Lewensohn R, Palti Y. Tumor treating fields (TTFields) delay DNA damage repair following radiation treatment of glioma cells. Radiat Oncol 2017; 12:206. [PMID: 29284495 PMCID: PMC5747183 DOI: 10.1186/s13014-017-0941-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 12/12/2017] [Indexed: 11/30/2022] Open
Abstract
Background Tumor Treating Fields (TTFields) are an anti-neoplastic treatment modality delivered via application of alternating electric fields using insulated transducer arrays placed directly on the skin in the region surrounding the tumor. A Phase 3 clinical trial has demonstrated the effectiveness of continuous TTFields application in patients with glioblastoma during maintenance treatment with Temozolomide. The goal of this study was to evaluate the efficacy of combining TTFields with radiation treatment (RT) in glioma cells. We also examined the effect of TTFields transducer arrays on RT distribution in a phantom model and the impact on rat skin toxicity. Methods The efficacy of TTFields application after induction of DNA damage by RT or bleomycin was tested in U-118 MG and LN-18 glioma cells. The alkaline comet assay was used to measure repair of DNA lesions. Repair of DNA double strand breaks (DSBs) were assessed by analyzing γH2AX or Rad51 foci. DNA damage and repair signaled by the activation pattern of phospho-ATM (pS1981) and phospho-DNA-PKcs (pS2056) was evaluated by immunoblotting. The absorption of the RT energy by transducer arrays was measured by applying RT through arrays placed on a solid-state phantom. Skin toxicities were tested in rats irradiated daily through the arrays with 2Gy (total dose of 20Gy). Results TTFields synergistically enhanced the efficacy of RT in glioma cells. Application of TTFields to irradiated cells impaired repair of irradiation- or chemically-induced DNA damage, possibly by blocking homologous recombination repair. Transducer arrays presence caused a minor reduction in RT intensity at 20 mm and 60 mm below the arrays, but led to a significant increase in RT dosage at the phantom surface jeopardizing the “skin sparing effect”. Nevertheless, transducer arrays placed on the rat skin during RT did not lead to additional skin reactions. Conclusions Administration of TTFields after RT increases glioma cells treatment efficacy possibly by inhibition of DNA damage repair. These preclinical results support the application of TTFields therapy immediately after RT as a viable regimen to enhance RT outcome. Phantom measurements and animal models imply that it may be possible to leave the transducer arrays in place during RT without increasing skin toxicities. Electronic supplementary material The online version of this article (10.1186/s13014-017-0941-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Petra Hååg
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden
| | | | | | | | - Kristina Viktorsson
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden
| | - Rolf Lewensohn
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden
| | | |
Collapse
|
731
|
Boyd NH, Walker K, Fried J, Hackney JR, McDonald PC, Benavides GA, Spina R, Audia A, Scott SE, Libby CJ, Tran AN, Bevensee MO, Griguer C, Nozell S, Gillespie GY, Nabors B, Bhat KP, Bar EE, Darley-Usmar V, Xu B, Gordon E, Cooper SJ, Dedhar S, Hjelmeland AB. Addition of carbonic anhydrase 9 inhibitor SLC-0111 to temozolomide treatment delays glioblastoma growth in vivo. JCI Insight 2017; 2:92928. [PMID: 29263302 DOI: 10.1172/jci.insight.92928] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 10/23/2017] [Indexed: 12/31/2022] Open
Abstract
Tumor microenvironments can promote stem cell maintenance, tumor growth, and therapeutic resistance, findings linked by the tumor-initiating cell hypothesis. Standard of care for glioblastoma (GBM) includes temozolomide chemotherapy, which is not curative, due, in part, to residual therapy-resistant brain tumor-initiating cells (BTICs). Temozolomide efficacy may be increased by targeting carbonic anhydrase 9 (CA9), a hypoxia-responsive gene important for maintaining the altered pH gradient of tumor cells. Using patient-derived GBM xenograft cells, we explored whether CA9 and CA12 inhibitor SLC-0111 could decrease GBM growth in combination with temozolomide or influence percentages of BTICs after chemotherapy. In multiple GBMs, SLC-0111 used concurrently with temozolomide reduced cell growth and induced cell cycle arrest via DNA damage in vitro. In addition, this treatment shifted tumor metabolism to a suppressed bioenergetic state in vivo. SLC-0111 also inhibited the enrichment of BTICs after temozolomide treatment determined via CD133 expression and neurosphere formation capacity. GBM xenografts treated with SLC-0111 in combination with temozolomide regressed significantly, and this effect was greater than that of temozolomide or SLC-0111 alone. We determined that SLC-0111 improves the efficacy of temozolomide to extend survival of GBM-bearing mice and should be explored as a treatment strategy in combination with current standard of care.
Collapse
Affiliation(s)
- Nathaniel H Boyd
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kiera Walker
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joshua Fried
- Department of Oncology, Southern Research Institute, Birmingham, Alabama, USA
| | - James R Hackney
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paul C McDonald
- Department of Integrative Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Gloria A Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Raffaella Spina
- Department of Neurological Surgery, Case Western University, Cleveland, Ohio, USA
| | - Alessandra Audia
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sarah E Scott
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Catherine J Libby
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anh Nhat Tran
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mark O Bevensee
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | - Burt Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Krishna P Bhat
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Eli E Bar
- Department of Neurological Surgery, Case Western University, Cleveland, Ohio, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bo Xu
- Department of Oncology, Southern Research Institute, Birmingham, Alabama, USA
| | - Emily Gordon
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Sara J Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Anita B Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
732
|
Advances in Brain Tumor Surgery for Glioblastoma in Adults. Brain Sci 2017; 7:brainsci7120166. [PMID: 29261148 PMCID: PMC5742769 DOI: 10.3390/brainsci7120166] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/24/2017] [Accepted: 12/13/2017] [Indexed: 01/18/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary intracranial neoplasia, and is characterized by its extremely poor prognosis. Despite maximum surgery, chemotherapy, and radiation, the histological heterogeneity of GBM makes total eradication impossible, due to residual cancer cells invading the parenchyma, which is not otherwise seen in radiographic images. Even with gross total resection, the heterogeneity and the dormant nature of brain tumor initiating cells allow for therapeutic evasion, contributing to its recurrence and malignant progression, and severely impacting survival. Visual delimitation of the tumor’s margins with common surgical techniques is a challenge faced by many surgeons. In an attempt to achieve optimal safe resection, advances in approaches allowing intraoperative analysis of cancer and non-cancer tissue have been developed and applied in humans resulting in improved outcomes. In addition, functional paradigms based on stimulation techniques to map the brain’s electrical activity have optimized glioma resection in eloquent areas such as the Broca’s, Wernike’s and perirolandic areas. In this review, we will elaborate on the current standard therapy for newly diagnosed and recurrent glioblastoma with a focus on surgical approaches. We will describe current technologies used for glioma resection, such as awake craniotomy, fluorescence guided surgery, laser interstitial thermal therapy and intraoperative mass spectrometry. Additionally, we will describe a newly developed tool that has shown promising results in preclinical experiments for brain cancer: optical coherence tomography.
Collapse
|
733
|
Stupp R, Taillibert S, Kanner A, Read W, Steinberg DM, Lhermitte B, Toms S, Idbaih A, Ahluwalia MS, Fink K, Di Meco F, Lieberman F, Zhu JJ, Stragliotto G, Tran DD, Brem S, Hottinger AF, Kirson ED, Lavy-Shahaf G, Weinberg U, Kim CY, Paek SH, Nicholas G, Burna J, Hirte H, Weller M, Palti Y, Hegi ME, Ram Z. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA 2017; 318:2306-2316. [PMID: 29260225 PMCID: PMC5820703 DOI: 10.1001/jama.2017.18718] [Citation(s) in RCA: 1659] [Impact Index Per Article: 207.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Tumor-treating fields (TTFields) is an antimitotic treatment modality that interferes with glioblastoma cell division and organelle assembly by delivering low-intensity alternating electric fields to the tumor. OBJECTIVE To investigate whether TTFields improves progression-free and overall survival of patients with glioblastoma, a fatal disease that commonly recurs at the initial tumor site or in the central nervous system. DESIGN, SETTING, AND PARTICIPANTS In this randomized, open-label trial, 695 patients with glioblastoma whose tumor was resected or biopsied and had completed concomitant radiochemotherapy (median time from diagnosis to randomization, 3.8 months) were enrolled at 83 centers (July 2009-2014) and followed up through December 2016. A preliminary report from this trial was published in 2015; this report describes the final analysis. INTERVENTIONS Patients were randomized 2:1 to TTFields plus maintenance temozolomide chemotherapy (n = 466) or temozolomide alone (n = 229). The TTFields, consisting of low-intensity, 200 kHz frequency, alternating electric fields, was delivered (≥ 18 hours/d) via 4 transducer arrays on the shaved scalp and connected to a portable device. Temozolomide was administered to both groups (150-200 mg/m2) for 5 days per 28-day cycle (6-12 cycles). MAIN OUTCOMES AND MEASURES Progression-free survival (tested at α = .046). The secondary end point was overall survival (tested hierarchically at α = .048). Analyses were performed for the intent-to-treat population. Adverse events were compared by group. RESULTS Of the 695 randomized patients (median age, 56 years; IQR, 48-63; 473 men [68%]), 637 (92%) completed the trial. Median progression-free survival from randomization was 6.7 months in the TTFields-temozolomide group and 4.0 months in the temozolomide-alone group (HR, 0.63; 95% CI, 0.52-0.76; P < .001). Median overall survival was 20.9 months in the TTFields-temozolomide group vs 16.0 months in the temozolomide-alone group (HR, 0.63; 95% CI, 0.53-0.76; P < .001). Systemic adverse event frequency was 48% in the TTFields-temozolomide group and 44% in the temozolomide-alone group. Mild to moderate skin toxicity underneath the transducer arrays occurred in 52% of patients who received TTFields-temozolomide vs no patients who received temozolomide alone. CONCLUSIONS AND RELEVANCE In the final analysis of this randomized clinical trial of patients with glioblastoma who had received standard radiochemotherapy, the addition of TTFields to maintenance temozolomide chemotherapy vs maintenance temozolomide alone, resulted in statistically significant improvement in progression-free survival and overall survival. These results are consistent with the previous interim analysis. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00916409.
Collapse
Affiliation(s)
- Roger Stupp
- Lou and Jean MalnatiBrain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center of Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Sophie Taillibert
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | | | - William Read
- University of California, San Diego
- Emory University, Atlanta, Georgia
| | | | - Benoit Lhermitte
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Steven Toms
- Geisinger Health System, Danville, Pennsylvania
| | - Ahmed Idbaih
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | | | - Karen Fink
- Baylor University Medical Center, Houston, Texas
| | | | - Frank Lieberman
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jay-Jiguang Zhu
- University of Texas Health Sciences Center at Houston
- Tufts Medical Center, Boston, Massachusetts
| | | | - David D. Tran
- Washington University Barnes-Jewish Hospital, St Louis, Missouri
| | - Steven Brem
- Moffitt Cancer Center, Tampa, Florida
- University of Pennsylvania, Philadelphia
| | - Andreas F. Hottinger
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | | | | | | | - Chae-Yong Kim
- Seoul National University Bundang Hospital, Seoul National University College of Medicine, Bundang, Korea
| | | | - Garth Nicholas
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jordi Burna
- Hospital Universitario de Bellvitge, Barcelona, Spain
| | - Hal Hirte
- Juravinski Cancer Centre, Hamilton, Ontario, Canada
| | - Michael Weller
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | | | - Monika E. Hegi
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Zvi Ram
- Tel Aviv Medical Center, Tel Aviv, Israel
| |
Collapse
|
734
|
Shi W, Blomain ES, Siglin J, Palmer JD, Dan T, Wang Y, Werner-Wasik M, Glass J, Kim L, Bar Ad V, Bhamidipati D, Evans JJ, Judy K, Farrell CJ, Andrews DW. Salvage fractionated stereotactic re-irradiation (FSRT) for patients with recurrent high grade gliomas progressed after bevacizumab treatment. J Neurooncol 2017; 137:171-177. [PMID: 29235052 DOI: 10.1007/s11060-017-2709-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 12/04/2017] [Indexed: 11/24/2022]
Abstract
Bevacizumab failure is a major clinical problem in the management of high grade gliomas (HGG), with a median overall survival (OS) of < 4 months. This study evaluated the feasibility and efficacy of fractionated stereotactic re-irradiation (FSRT) for patients progressed after Bevacizumab treatment. Retrospective review was conducted of 36 patients treated with FSRT after progression on bevacizumab. FSRT was most commonly delivered in 3.5 Gy fractions to a total dose of 35 Gy. Survival from initial diagnosis, as well as from recurrence and re-irradiation, were utilized as study endpoints. Univariate and multivariate analysis was performed. The median time from initial bevacizumab treatment to FSRT was 8.5 months. The median plan target volume for FSRT was 27.5 cc. The median OS from FSRT was 4.8 months. FSRT treatment was well tolerated with no grade 3 or higher toxicity. Favorable outcomes were observed in patients with recurrent HGG who received salvage FSRT after bevacizumab failure. The treatment was well tolerated. Prospective study is warranted to further evaluate the efficacy of salvage FSRT for selected patients with recurrent HGG amenable to FSRT, who had failed bevacizumab treatment.
Collapse
Affiliation(s)
- Wenyin Shi
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Erik S Blomain
- Jefferson College of Population Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joshua Siglin
- Department of Radiation Oncology, University of Pittsburgh Medical Center, Altoona Cancer Center, Altoona, PA, USA
| | - Joshua D Palmer
- Department of Radiation Oncology, Ohio State University, Columbus, OH, USA
| | - Tu Dan
- Department of Radiation Oncology, University of Texas Southwestern, Dallas, TX, USA
| | - Yang Wang
- Cyberknife Center, Huashan Hospital Pudong, Fudan University, Shanghai, China
| | - Maria Werner-Wasik
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jon Glass
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lyndon Kim
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Voichita Bar Ad
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Deepak Bhamidipati
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - James J Evans
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kevin Judy
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christopher J Farrell
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - David W Andrews
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
735
|
Ge J, Hu W, Zhou H, Yu J, Sun C, Chen W. Ubiquitin carboxyl-terminal hydrolase isozyme L5 inhibits human glioma cell migration and invasion via downregulating SNRPF. Oncotarget 2017; 8:113635-113649. [PMID: 29371935 PMCID: PMC5768352 DOI: 10.18632/oncotarget.23071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/14/2017] [Indexed: 12/14/2022] Open
Abstract
Ubiquitin C-terminal Hydrolase-L5 (UCH-L5/UCH37), a member of the deubiquitinases (DUBs), suppresses protein degeneration via removing ubiquitin from the distal subunit of the polyubiquitin chain. The activity of UCH-L5 is enhanced when UCH-L5 combines with proteasome 19S regulatory subunit by Rpn13/Admr1 receptor and inhibited when UCH-L5 interacts with NFRKB. But the role of UCH-L5 in gliomas remains unknown. In this study, analysis of 19 frozen and 51 paraffin-embedded clinic pathological cases showed that UCH-L5 expression in glioma tissues was lower than normal brain tissues. In vitro, we found that UCH-L5 could inhibit migration and invasion of U87MG and U251 cells. It has been reported that the expression of SNRPN, SNRPF, and CKLF was abnormal in gliomas or other tumors. We also found that SNRPF-siRNA, SNRPN-siRNA and CKLF-siRNA could inhibit migration and invasion of U87MG cells. And knockdown of UCH-L5 expression improved both mRNA expression and protein level of SNRPF. The relationship between UCH-L5 and SNRPF was further confirmed in 293T cells. Our study showed that UCH-L5 could inhibit migration and invasion of glioma cells via down regulating expression of SNRPF. And the above findings suggest that UCH-L5 may inhibit occurrence and metastasis of gliomas.
Collapse
Affiliation(s)
- Jiafeng Ge
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weiwei Hu
- Department of Neurosurgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hui Zhou
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Juan Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chongran Sun
- Department of Neurosurgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weilin Chen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Department of Immunology, Shenzhen University School of Medicine, Shenzhen 518060, China
| |
Collapse
|
736
|
Jiang S, Hill K, Patel D, Waldeck AR, Botteman M, Aly A, Norden AD. Direct medical costs of treatment in newly-diagnosed high-grade glioma among commercially insured US patients. J Med Econ 2017; 20:1237-1243. [PMID: 28777020 DOI: 10.1080/13696998.2017.1364258] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
AIM This analysis assessed the direct medical costs of newly-diagnosed, temozolomide (TMZ)-treated glioblastoma (GBM) from the perspective of a US commercial setting. MATERIALS AND METHODS The analysis included subjects identified from the IMS PharMetrics LifeLink Plus™ claims database from January 1, 2008 to August 31, 2014 who were ≥18 years of age, had ≥1 malignant brain cancer diagnosis, had brain surgery ≤90 days prior to TMZ initiation, had TMZ treatment, and were continuously enrolled for ≥12 months pre-diagnosis and ≥1 month post-diagnosis. Per-patient per-month (PPPM) and cumulative costs from 3 months pre-diagnosis to various post-diagnosis follow-up time points were calculated. Multivariable analyses were used to estimate adjusted mean cost and identify contributors of cost. RESULTS The study included 2,921 subjects (median age = 56 years; 60% male). After diagnosis, the median (interquartile range, IQR) number of inpatient, emergency department, and outpatient visits were 2 (1-4), 1 (1-3), and 19 (13-27); median (IQR) length of stay per hospitalization was 5 (3-9) days. Mean total cumulative costs per patient from 3 months pre-diagnosis to 12 months and to 5 years post-diagnosis were $201,749 (197,490-206,024) and $268,031 (262,877-274,416). Mean (SD) PPPM costs were $818 (1,128) and $7,394 (8,676) pre- and post-GBM diagnosis, respectively. The variables most predictive of cumulative costs included radiation therapy (+$81,732), ≥2 weeks of hospitalization (+$49,629), and ≥7 MRI scans (+$40,105). CONCLUSIONS The direct medical costs of newly-diagnosed, TMZ-treated GBM in commercially insured patients are substantial, with estimated total cumulative costs of $268,031.
Collapse
Affiliation(s)
- Shan Jiang
- a Pharmerit International , Bethesda , MD , USA
| | - Kala Hill
- b Celldex Therapeutics Inc. , Hampton , NJ , USA
| | - Dipen Patel
- a Pharmerit International , Bethesda , MD , USA
| | | | | | - Abdalla Aly
- a Pharmerit International , Bethesda , MD , USA
| | - Andrew D Norden
- c Dana-Farber/Brigham and Women's Cancer Center , Boston , MA , USA
| |
Collapse
|
737
|
Lukas RV, Mrugala MM. Pivotal therapeutic trials for infiltrating gliomas and how they affect clinical practice. Neurooncol Pract 2017; 4:209-219. [PMID: 31385973 PMCID: PMC6655416 DOI: 10.1093/nop/npw016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Indexed: 11/15/2022] Open
Abstract
The therapeutic landscape of the management of low- and high-grade infiltrating gliomas continues to evolve. Daily clinical decision making in neuro-oncology clinics across the US is frequently challenging, especially for anaplastic and low grade primary brain tumors. The focus of this review is centered on treatments which are approved by the FDA and/or featured in the NCCN Guidelines. Systemic therapy trials using a variety of agents such as temozolomide, bevacizumab, and procarbazine, lomustine, vincristine (PCV), and lastly trials of local therapies including surgical trials using carmustine impregnated wafers as well as trials investigating the administration of tumor treating fields are evaluated. Pivotal trials on the treatment of the primary brain tumors are discussed in detail along with associated correlative studies.
Collapse
Affiliation(s)
- Rimas V Lukas
- University of Chicago, Department of Neurology (R.V.L.); University of Washington, Department of Neurology (M.M.M.)
| | | |
Collapse
|
738
|
Adeberg S, Harrabi SB, Verma V, Bernhardt D, Grau N, Debus J, Rieken S. Treatment of meningioma and glioma with protons and carbon ions. Radiat Oncol 2017; 12:193. [PMID: 29195506 PMCID: PMC5710063 DOI: 10.1186/s13014-017-0924-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/14/2017] [Indexed: 11/10/2022] Open
Abstract
The rapid rise of particle therapy across the world necessitates evidence to justify its ever-increasing utilization. This narrative review summarizes the current status of these technologies on treatment of both meningiomas and gliomas, the most common benign and malignant primary brain tumors, respectively. Proton beam therapy (PBT) for meningiomas displays high rates of long-term local control, low rates of symptomatic deterioration, along with the potential for safe dose-escalation in select (but not necessarily routine) cases. PBT is also associated with low adverse events and maintenance of functional outcomes, which have implications for quality of life and cost-effectiveness measures going forward. Data on carbon ion radiation therapy (CIRT) are limited; existing series describe virtually no high-grade toxicities and high local control. Regarding the few available data on low-grade gliomas, PBT provides opportunities to dose-escalate while affording no increase of severe toxicities, along with maintaining appropriate quality of life. Although dose-escalation for low-grade disease has been less frequently performed than for glioblastoma, PBT and CIRT continue to be utilized for the latter, and also have potential for safer re-irradiation of high-grade gliomas. For both neoplasms, the impact of superior dosimetric profiles with endpoints such as neurocognitive decline and neurologic funcionality, are also discussed to the extent of requiring more data to support the utility of particle therapy. Caveats to these data are also described, such as the largely retrospective nature of the available studies, patient selection, and heterogeneity in patient population as well as treatment (including mixed photon/particle treatment). Nevertheless, multiple prospective trials (which may partially attenuate those concerns) are also discussed. In light of the low quantity and quality of available data, major questions remain regarding economic concerns as well.
Collapse
Affiliation(s)
- Sebastian Adeberg
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany. .,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,Heidelberg Ion-Beam Therapy Center, Im Neuenheimer Feld 450, 69120, Heidelberg, Germany. .,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), 69120, Heidelberg, Germany.
| | - Semi B Harrabi
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Im Neuenheimer Feld 450, 69120, Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), 69120, Heidelberg, Germany
| | - Vivek Verma
- Department of Radiation Oncology, University of Nebraska Medical Center, 505 S 45th Street, Omaha, NE, 68106, USA
| | - Denise Bernhardt
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Im Neuenheimer Feld 450, 69120, Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), 69120, Heidelberg, Germany
| | - Nicole Grau
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), 69120, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Im Neuenheimer Feld 450, 69120, Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), 69120, Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Im Neuenheimer Feld 450, 69120, Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), 69120, Heidelberg, Germany
| |
Collapse
|
739
|
Gusyatiner O, Hegi ME. Glioma epigenetics: From subclassification to novel treatment options. Semin Cancer Biol 2017; 51:50-58. [PMID: 29170066 DOI: 10.1016/j.semcancer.2017.11.010] [Citation(s) in RCA: 355] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/26/2022]
Abstract
Gliomas are the most common malignant primary brain tumors, of which glioblastoma is the most malignant form (WHO grade IV), and notorious for treatment resistance. Over the last decade mutations in epigenetic regulator genes have been identified as key drivers of subtypes of gliomas with distinct clinical features. Most characteristic are mutations in IDH1 or IDH2 in lower grade gliomas, and histone 3 mutations in pediatric high grade gliomas that are also associated with characteristic DNA methylation patterns. Furthermore, in adult glioblastoma patients epigenetic silencing of the DNA repair gene MGMT by promoter methylation is predictive for benefit from alkylating agent therapy. These epigenetic alterations are used as biomarkers and play a central role for classification of gliomas (WHO 2016) and treatment decisions. Here we review the pivotal role of epigenetic alterations in the etiology and biology of gliomas. We summarize the complex interactions between "driver" mutations, DNA methylation, histone post-translational modifications, and overall chromatin organization, and how they inform current efforts of testing epigenetic compounds and combinations in preclinical and clinical studies.
Collapse
Affiliation(s)
- Olga Gusyatiner
- Laboratory of Brain Tumor Biology and Genetics, Neuroscience Research Center and Service of Neurosurgery, Lausanne University Hospital, 1066 Epalinges, Switzerland
| | - Monika E Hegi
- Laboratory of Brain Tumor Biology and Genetics, Neuroscience Research Center and Service of Neurosurgery, Lausanne University Hospital, 1066 Epalinges, Switzerland.
| |
Collapse
|
740
|
Zorniak M, Clark PA, Umlauf BJ, Cho Y, Shusta EV, Kuo JS. Yeast display biopanning identifies human antibodies targeting glioblastoma stem-like cells. Sci Rep 2017; 7:15840. [PMID: 29158489 PMCID: PMC5696472 DOI: 10.1038/s41598-017-16066-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 11/07/2017] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma stem-like cells (GSC) are hypothesized to evade current therapies and cause tumor recurrence, contributing to poor patient survival. Existing cell surface markers for GSC are developed from embryonic or neural stem cell systems; however, currently available GSC markers are suboptimal in sensitivity and specificity. We hypothesized that the GSC cell surface proteome could be mined with a yeast display antibody library to reveal novel immunophenotypes. We isolated an extensive collection of antibodies that were differentially selective for GSC. A single domain antibody VH-9.7 showed selectivity for five distinct patient-derived GSC lines and visualized orthotopic GBM xenografts in vivo after conjugation with a near-infrared dye. These findings demonstrate a previously unexplored high-throughput strategy for GSC-selective antibody discovery, to aid in GSC isolation, diagnostic imaging, and therapeutic targeting.
Collapse
Affiliation(s)
- Michael Zorniak
- Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA.,Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA
| | - Paul A Clark
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA
| | - Benjamin J Umlauf
- Department of Chemical and Biological Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA
| | - Yongku Cho
- Department of Chemical and Biological Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA. .,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA.
| | - John S Kuo
- Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA. .,Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA. .,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53792-8660, USA.
| |
Collapse
|
741
|
Migliorini D, Dietrich PY, Stupp R, Linette GP, Posey AD, June CH. CAR T-Cell Therapies in Glioblastoma: A First Look. Clin Cancer Res 2017; 24:535-540. [PMID: 29158268 DOI: 10.1158/1078-0432.ccr-17-2871] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/08/2017] [Accepted: 11/16/2017] [Indexed: 11/16/2022]
Abstract
Glioblastoma is an aggressive malignancy with a poor prognosis. The current standard of care for newly diagnosed glioblastoma patients includes surgery to the extent, temozolomide combined with radiotherapy, and alternating electric fields therapy. After recurrence, there is no standard therapy and survival is less than 9 months. Recurrent glioblastoma offers a unique opportunity to investigate new treatment approaches in a malignancy known for remarkable genetic heterogeneity, an immunosuppressive microenvironment, and a partially permissive anatomic blood-brain barrier. Results from three first-in-man chimeric antigen receptor (CAR) T-cell trials targeting IL13Rα2, Her2/CMV, and EGFRvIII have recently been reported. Each one of these trials addresses important questions, such as T-cell trafficking to CNS, engraftment and persistence, tumor microenvironment remodeling, and monitoring of glioma response to CAR T cells. Objective radiologic responses have been reported. Here, we discuss and summarize the results of these trials and suggest opportunities for the field. Clin Cancer Res; 24(3); 535-40. ©2017 AACR.
Collapse
Affiliation(s)
- Denis Migliorini
- Center for Cellular Immunotherapies and Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Roger Stupp
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Gerald P Linette
- Center for Cellular Immunotherapies and Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Avery D Posey
- Center for Cellular Immunotherapies and Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carl H June
- Center for Cellular Immunotherapies and Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
742
|
Initial psycho-oncological counselling in neuro-oncology: analysis of topics and needs of brain tumour patients. J Neurooncol 2017; 136:505-514. [PMID: 29147859 DOI: 10.1007/s11060-017-2670-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 11/11/2017] [Indexed: 10/18/2022]
Abstract
Diagnosis of a brain tumour is associated with a tremendous disruption of emotional, physical and social well-being. Due to the complexity of the disease and the affection of the central organ, the brain, brain tumour patients differ from other cancer patients. The purpose of this study was to evaluate the concerns and burdens presented by brain tumour patients during their initial psycho-oncological consultation. We performed a retrospective analysis of 53 patients with the diagnosis of either benign or malignant brain tumour, seeking counsel by a neurosurgeon specialised in psycho-oncology. We performed a thematic analysis of the interviews at first consultation identifying themes and patterns and created thematic categories. The main concerns of the patients presented during the first consultations were psychological problems, reported by 40 patients (75.5%). Death and dying was mentioned by more than half of the patients (n = 30, 56.6%). In addition, 62.3% of the patients (n = 33) asked for information regarding the medical treatment and diagnosis. With our study, we created greater awareness of the psychological needs of brain tumour patients in order to define treatment strategies for this important aspect of disease. We showed that there is a need for patients to talk about death even during the initial consultation. Psycho-oncologist in a neuro-oncological setting should be prepared for topics like that and should have a neurosurgical background or collaborate with members of the surgical team in order to provide the patients with medical details and to better understand the impact of the disease.
Collapse
|
743
|
Wick W, Kessler T. Drug Repositioning Meets Precision in Glioblastoma. Clin Cancer Res 2017; 24:256-258. [PMID: 29133572 DOI: 10.1158/1078-0432.ccr-17-2989] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 10/28/2017] [Accepted: 11/07/2017] [Indexed: 11/16/2022]
Abstract
Glioblastoma has a gigantic unmet medical need. Molecular knowledge has evolved substantially, including data on clonal selection with progression. Past trials for all-comers may have produced false negative results. Molecular precision at progression needs workup of new tissue, and revisiting drugs with a focus on brain tumor penetration may yield surprises. Clin Cancer Res; 24(2); 256-8. ©2017 AACRSee related article by Byron et al., p. 295.
Collapse
Affiliation(s)
- Wolfgang Wick
- Neurology Clinic, University of Heidelberg, Heidelberg, Germany. .,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tobias Kessler
- Neurology Clinic, University of Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
744
|
Santos JG, Da Cruz WMS, Schönthal AH, Salazar MD, Fontes CAP, Quirico-Santos T, Da Fonseca CO. Efficacy of a ketogenic diet with concomitant intranasal perillyl alcohol as a novel strategy for the therapy of recurrent glioblastoma. Oncol Lett 2017; 15:1263-1270. [PMID: 29391903 DOI: 10.3892/ol.2017.7362] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 06/09/2017] [Indexed: 12/20/2022] Open
Abstract
It has been hypothesized that persistent ketotic hypoglycemia represents a potential therapeutic strategy against high-grade gliomas. Perillyl alcohol (POH) is a non-toxic, naturally-occurring, hydroxylated monoterpene that exhibits cytotoxicity against temozolomide-resistant glioma cells, regardless of O6-methylguanine-methyltransferase promoter methylation status. The present study aimed to evaluate the toxicity and therapeutic efficacy of intranasal POH when administered in combination with a ketogenic diet (KD) program for the treatment of patients with recurrent glioblastoma. The 32 enrolled patients were divided into two groups, KD or standard diet, with intranasal POH treatment (n=17 and n=15, respectively). The nutritional status and anthropometric parameters of the patients were measured. Patients that adhered to the KD maintained a strict dietary regimen, in addition to receiving 55 mg POH four times daily, in an uninterrupted administration schedule for three months. Neurological examination and magnetic resonance imaging analysis were used to monitor disease progression. A total of 9/17 patients in the KD group survived and maintained compliance with the KD. After three months of well-tolerated treatment, a partial response (PR) was observed for 77.8% (7/9) of the patients, stable disease (SD) in 11.1% (1/9) and 11.1% (1/9) presented with progressive disease (PD). Among the patients assigned to the standard diet group, the PR rate was 25% (2/8 patients), SD 25% (2/8) and PD 50% (4/8 patients). The patients assigned to the KD group presented with reduced serum lipid levels and decreased low-density lipoprotein cholesterol levels. These results are encouraging and suggest that KD associated with intranasal POH may represent a viable option as an adjunct therapy for recurrent GBM.
Collapse
Affiliation(s)
- Juliana Guimarães Santos
- Graduate Program in Medical Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro 24033-900, Brazil
| | | | - Axel H Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | | | - Cristina Asvolinsque Pantaleão Fontes
- Service of Radiology, Department of Radiology, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi, Rio de Janeiro 24033-900, Brazil
| | - Thereza Quirico-Santos
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Rio de Janeiro 24210-130, Brazil
| | - Clovis Orlando Da Fonseca
- Service of Neurosurgery, Department of General and Specialized Surgery, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi, Rio de Janeiro 24033-900, Brazil
| |
Collapse
|
745
|
Xu H, Chen J, Xu H, Qin Z. Geographic Variations in the Incidence of Glioblastoma and Prognostic Factors Predictive of Overall Survival in US Adults from 2004-2013. Front Aging Neurosci 2017; 9:352. [PMID: 29163134 PMCID: PMC5681990 DOI: 10.3389/fnagi.2017.00352] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 10/17/2017] [Indexed: 12/22/2022] Open
Abstract
Objective: The purpose of this study was to evaluate variations in the regional incidence of glioblastoma in US adults in 2004-2013. Study Design and Setting: We evaluated 24,262 patients with primary glioblastoma. Data were categorized based on geographic regions that included different SEER registry sites as follows: (1) Northeast: Connecticut, New Jersey (3,977 patients); (2) South: Kentucky, Louisiana, Metropolitan Atlanta, Rural Georgia, Greater Georgia (excluding AT and RG) (5,212 patients); (3) North Central: Metropolitan Detroit, Iowa (2,320 patients); (4) West: Hawaii, New Mexico, Seattle (Puget Sound), Utah, San Francisco-Oakland SMSA, San Jose-Monterey, Los Angeles, Greater California (excluding SF, LA, and SJ), Alaska (12,753 patients). Results: Statistically significant differences in the rates of overall patient survival (P < 0.001) and the incidence of glioblastoma (24.31, 22.6, 20.35, 15.03 per 100,000/year in the South, Northeast, West, North Central regions, respectively) were identified between geographic regions. Multivariate Cox regression analysis demonstrated that overall survival was better in patients of Asian or Pacific Islander race. In addition, age, registry site, marital status, tumor laterality, histological classification, the extent of disease, tumor size, tumor extension, and treatment methods were identified as significant prognostic factors. Conclusion: Glioblastoma incidence is geographic region and race/ethnicity-dependent.
Collapse
Affiliation(s)
| | | | | | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
746
|
Harford-Wright E, Andre-Gregoire G, Jacobs KA, Treps L, Le Gonidec S, Leclair HM, Gonzalez-Diest S, Roux Q, Guillonneau F, Loussouarn D, Oliver L, Vallette FM, Foufelle F, Valet P, Davenport AP, Glen RC, Bidere N, Gavard J. Pharmacological targeting of apelin impairs glioblastoma growth. Brain 2017; 140:2939-2954. [PMID: 29053791 PMCID: PMC5841205 DOI: 10.1093/brain/awx253] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 08/05/2017] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma are highly aggressive brain tumours that are associated with an extremely poor prognosis. Within these tumours exists a subpopulation of highly plastic self-renewing cancer cells that retain the ability to expand ex vivo as tumourspheres, induce tumour growth in mice, and have been implicated in radio- and chemo-resistance. Although their identity and fate are regulated by external cues emanating from endothelial cells, the nature of such signals remains unknown. Here, we used a mass spectrometry proteomic approach to characterize the factors released by brain endothelial cells. We report the identification of the vasoactive peptide apelin as a central regulator for endothelial-mediated maintenance of glioblastoma patient-derived cells with stem-like properties. Genetic and pharmacological targeting of apelin cognate receptor abrogates apelin- and endothelial-mediated expansion of glioblastoma patient-derived cells with stem-like properties in vitro and suppresses tumour growth in vivo. Functionally, selective competitive antagonists of apelin receptor were shown to be safe and effective in reducing tumour expansion and lengthening the survival of intracranially xenografted mice. Therefore, the apelin/apelin receptor signalling nexus may operate as a paracrine signal that sustains tumour cell expansion and progression, suggesting that apelin is a druggable factor in glioblastoma.
Collapse
Affiliation(s)
- Elizabeth Harford-Wright
- CRCINA, Inserm, Team SOAP, CNRS, Universite de Nantes, Nantes, France.,Institut Cochin, Team SOAP, Inserm, CNRS, Universite Paris Descartes, Paris, France
| | | | - Kathryn A Jacobs
- CRCINA, Inserm, Team SOAP, CNRS, Universite de Nantes, Nantes, France
| | - Lucas Treps
- Institut Cochin, Team SOAP, Inserm, CNRS, Universite Paris Descartes, Paris, France
| | | | - Heloise M Leclair
- CRCINA, Inserm, Team SOAP, CNRS, Universite de Nantes, Nantes, France.,Institut Cochin, Team SOAP, Inserm, CNRS, Universite Paris Descartes, Paris, France
| | - Sara Gonzalez-Diest
- CRCINA, Inserm, Team SOAP, CNRS, Universite de Nantes, Nantes, France.,Institut Cochin, Team SOAP, Inserm, CNRS, Universite Paris Descartes, Paris, France
| | - Quentin Roux
- CRCINA, Inserm, Team SOAP, CNRS, Universite de Nantes, Nantes, France
| | | | - Delphine Loussouarn
- Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France.,CRCINA, Inserm, Universite de Nantes, Nantes, France
| | - Lisa Oliver
- Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France.,CRCINA, Inserm, Universite de Nantes, Nantes, France
| | - François M Vallette
- CRCINA, Inserm, Universite de Nantes, Nantes, France.,Institut de Cancérologie de l'Ouest, René Gauducheau, St Herblain, France
| | - Fabienne Foufelle
- Centre de Recherches des Cordeliers, Inserm, Universite Paris Descartes, Paris, France
| | - Philippe Valet
- I2MC, Inserm, Universite Paul Sabatier, Toulouse, France
| | - Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Robert C Glen
- The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK.,Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, UK
| | - Nicolas Bidere
- CRCINA, Inserm, Team SOAP, CNRS, Universite de Nantes, Nantes, France.,Institut Cochin, Team SOAP, Inserm, CNRS, Universite Paris Descartes, Paris, France
| | - Julie Gavard
- CRCINA, Inserm, Team SOAP, CNRS, Universite de Nantes, Nantes, France.,Institut Cochin, Team SOAP, Inserm, CNRS, Universite Paris Descartes, Paris, France
| |
Collapse
|
747
|
Abstract
Glioblastoma is the most aggressive brain tumor and, even with the current multimodal therapy, is an invariably lethal cancer with a life expectancy that depends on the tumor subtype but, even in the most favorable cases, rarely exceeds 2 years. Epigenetic factors play an important role in gliomagenesis, are strong predictors of outcome, and are important determinants for the resistance to radio- and chemotherapy. The latest addition to the epigenetic machinery is the noncoding RNA (ncRNA), that is, RNA molecules that are not translated into a protein and that exert their function by base pairing with other nucleic acids in a reversible and nonmutational mode. MicroRNAs (miRNA) are a class of ncRNA of about 22 bp that regulate gene expression by binding to complementary sequences in the mRNA and silence its translation into proteins. MicroRNAs reversibly regulate transcription through nonmutational mechanisms; accordingly, they can be considered as epigenetic effectors. In this review, we will discuss the role of miRNA in glioma focusing on their role in drug resistance and on their potential applications in the therapy of this tumor.
Collapse
|
748
|
Hauser A, Dutta SW, Showalter TN, Sheehan JP, Grover S, Trifiletti DM. Impact of academic facility type and volume on post-surgical outcomes following diagnosis of glioblastoma. J Clin Neurosci 2017; 47:103-110. [PMID: 29113851 DOI: 10.1016/j.jocn.2017.10.087] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/23/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To identify if facility type and/or facility volume impact overall survival (OS) following diagnosis of glioblastoma (GBM). We also sought to compare early post-surgical outcomes based on these factors. METHODS The National Cancer Database was queried for patients with GBM diagnosed from 2004 to 2013 with known survival. Patients were grouped based on facility type and facility volume. Multivariable analyses were performed to investigate factors associated OS following diagnosis and Chi-square tests were used to compare early post-surgical outcomes. RESULTS 89,839 patients met inclusion criteria. Factors associated with improved OS on multivariable analysis included younger patient age, female gender, race, lower comorbidity score, higher performance score, smaller tumor size, unifocal tumors, MGMT hypermethylation, fully resected tumors, radiotherapy, and chemotherapy (each p < .001). Also, OS was improved among patients treated at centers averaging at least 30.2 cases per year (HR 0.948, compared to <7.4 cases/year, p < .001), and patients treated at Academic/Research programs had improved survival compared to those treated at Comprehensive Community Cancer programs (HR 1.069, p < .001) and Integrated Network Cancer programs (HR 1.126, p < .001). Similarly, Academic/Research programs and high volume centers demonstrated improved 30- and 90-day morality as well as 30-day readmission rates (p < .001). CONCLUSIONS This study suggests that patients treated in Academic/Research programs and high patient-volume centers have increased survival and more favorable early-postsurgical outcomes. The extent to which differences in patient populations, socioeconomic factors, and/or provider expertise play into this cause will be areas of future research.
Collapse
Affiliation(s)
- Alan Hauser
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA, USA
| | - Sunil W Dutta
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA, USA
| | - Timothy N Showalter
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA, USA
| | - Jason P Sheehan
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA, USA; Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Surbhi Grover
- Department of Radiation Oncology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
749
|
Beba Abadal K, Walsh MA, Yachnis AT, Tran DD, Ghiaseddin AP. Eleven Month Progression–Free Survival on Vemurafenib Monotherapy in a Patient With Recurrent and Metastatic BRAF V600E–Mutated Glioblastoma WHO Grade 4. JCO Precis Oncol 2017; 1:1-5. [DOI: 10.1200/po.17.00055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
750
|
Filley AC, Henriquez M, Dey M. Recurrent glioma clinical trial, CheckMate-143: the game is not over yet. Oncotarget 2017; 8:91779-91794. [PMID: 29207684 PMCID: PMC5710964 DOI: 10.18632/oncotarget.21586] [Citation(s) in RCA: 270] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/08/2017] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma (GBM) is the most common, and aggressive, primary brain tumor in adults. With a median patient survival of less than two years, GBM represents one of the biggest therapeutic challenges of the modern era. Even with the best available treatment, recurrence rates are nearly 100% and therapeutic options at the time of relapse are extremely limited. Nivolumab, an anti-programmed cell death-1 (PD-1) monoclonal antibody, has provided significant clinical benefits in the treatment of various advanced cancers and represented a promising therapy for primary and recurrent GBM. CheckMate 143 (NCT 02017717) was the first large randomized clinical trial of PD pathway inhibition in the setting of GBM, including a comparison of nivolumab and the anti-VEGF antibody, bevacizumab, in the treatment of recurrent disease. However, preliminary results, recently announced in a WFNOS 2017 abstract, demonstrated a failure of nivolumab to prolong overall survival of patients with recurrent GBM, and this arm of the trial was prematurely closed. In this review, we discuss the basic concepts underlying the rational to target PD pathway in GBM, address implications of using immune checkpoint inhibitors in central nervous system malignancies, provide a rationale for possible reasons contributing to the failure of nivolumab to prolong survival in patients with recurrent disease, and analyze the future role of immune checkpoint inhibitors in the treatment of GBM.
Collapse
Affiliation(s)
- Anna C. Filley
- Department of Neurosurgery, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Mario Henriquez
- Department of Neurosurgery, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Mahua Dey
- Department of Neurosurgery, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| |
Collapse
|