601
|
Chandana S, Babiker HM, Mahadevan D. Therapeutic trends in pancreatic ductal adenocarcinoma (PDAC). Expert Opin Investig Drugs 2018; 28:161-177. [DOI: 10.1080/13543784.2019.1557145] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Sreenivasa Chandana
- Phase I program, START Midwest, Grand Rapids, MI, USA
- Department of Gastrointestinal Medical Oncology, Cancer and Hematology Centers of Western Michigan, Grand Rapids, MI, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Hani M. Babiker
- Early Phase Therapeutics Program, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Daruka Mahadevan
- Early Phase Therapeutics Program, University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
602
|
Latouche EL, Arena CB, Ivey JW, Garcia PA, Pancotto TE, Pavlisko N, Verbridge SS, Davalos RV, Rossmeisl JH. High-Frequency Irreversible Electroporation for Intracranial Meningioma: A Feasibility Study in a Spontaneous Canine Tumor Model. Technol Cancer Res Treat 2018; 17:1533033818785285. [PMID: 30071778 PMCID: PMC6077896 DOI: 10.1177/1533033818785285] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
High-frequency irreversible electroporation is a nonthermal method of tissue ablation
that uses bursts of 0.5- to 2.0-microsecond bipolar electric pulses to permeabilize cell
membranes and induce cell death. High-frequency irreversible electroporation has potential
advantages for use in neurosurgery, including the ability to deliver pulses without
inducing muscle contraction, inherent selectivity against malignant cells, and the
capability of simultaneously opening the blood–brain barrier surrounding regions of
ablation. Our objective was to determine whether high-frequency irreversible
electroporation pulses capable of tumor ablation could be delivered to dogs with
intracranial meningiomas. Three dogs with intracranial meningiomas were treated.
Patient-specific treatment plans were generated using magnetic resonance imaging-based
tissue segmentation, volumetric meshing, and finite element modeling. Following tumor
biopsy, high-frequency irreversible electroporation pulses were stereotactically delivered
in situ followed by tumor resection and morphologic and volumetric
assessments of ablations. Clinical evaluations of treatment included pre- and
posttreatment clinical, laboratory, and magnetic resonance imaging examinations and
adverse event monitoring for 2 weeks posttreatment. High-frequency irreversible
electroporation pulses were administered successfully in all patients. No adverse events
directly attributable to high-frequency irreversible electroporation were observed.
Individual ablations resulted in volumes of tumor necrosis ranging from 0.25 to 1.29
cm3. In one dog, nonuniform ablations were observed, with viable tumor cells
remaining around foci of intratumoral mineralization. In conclusion, high-frequency
irreversible electroporation pulses can be delivered to brain tumors, including areas
adjacent to critical vasculature, and are capable of producing clinically relevant volumes
of tumor ablation. Mineralization may complicate achievement of complete tumor
ablation.
Collapse
Affiliation(s)
| | | | - Jill W Ivey
- 2 Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University School of Biomedical Engineering, Blacksburg, VA, USA
| | | | - Theresa E Pancotto
- 3 Veterinary and Comparative Neuro-oncology Laboratory, Virginia Tech, Blacksburg, VA, USA.,4 Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Noah Pavlisko
- 3 Veterinary and Comparative Neuro-oncology Laboratory, Virginia Tech, Blacksburg, VA, USA.,4 Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Scott S Verbridge
- 2 Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University School of Biomedical Engineering, Blacksburg, VA, USA
| | - Rafael V Davalos
- 2 Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University School of Biomedical Engineering, Blacksburg, VA, USA
| | - John H Rossmeisl
- 2 Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest University School of Biomedical Engineering, Blacksburg, VA, USA.,3 Veterinary and Comparative Neuro-oncology Laboratory, Virginia Tech, Blacksburg, VA, USA.,4 Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
603
|
Chang E, Patel CB, Pohling C, Young C, Song J, Flores TA, Zeng Y, Joubert LM, Arami H, Natarajan A, Sinclair R, Gambhir SS. Tumor treating fields increases membrane permeability in glioblastoma cells. Cell Death Discov 2018; 4:113. [PMID: 30534421 PMCID: PMC6281619 DOI: 10.1038/s41420-018-0130-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma is the most common yet most lethal of primary brain cancers with a one-year post-diagnosis survival rate of 65% and a five-year survival rate of barely 5%. Recently the U.S. Food and Drug Administration approved a novel fourth approach (in addition to surgery, radiation therapy, and chemotherapy) to treating glioblastoma; namely, tumor treating fields (TTFields). TTFields involves the delivery of alternating electric fields to the tumor but its mechanisms of action are not fully understood. Current theories involve TTFields disrupting mitosis due to interference with proper mitotic spindle assembly. We show that TTFields also alters cellular membrane structure thus rendering it more permeant to chemotherapeutics. Increased membrane permeability through the imposition of TTFields was shown by several approaches. For example, increased permeability was indicated through increased bioluminescence with TTFields exposure or with the increased binding and ingress of membrane-associating reagents such as Dextran-FITC or ethidium D or with the demonstration by scanning electron microscopy of augmented number and sizes of holes on the cellular membrane. Further investigations showed that increases in bioluminescence and membrane hole production with TTFields exposure disappeared by 24 h after cessation of alternating electric fields thus demonstrating that this phenomenom is reversible. Preliminary investigations showed that TTFields did not induce membrane holes in normal human fibroblasts thus suggesting that the phenomenom was specific to cancer cells. With TTFields, we present evidence showing augmented membrane accessibility by compounds such as 5-aminolevulinic acid, a reagent used intraoperatively to delineate tumor from normal tissue in glioblastoma patients. In addition, this mechanism helps to explain previous reports of additive and synergistic effects between TTFields and other chemotherapies. These findings have implications for the design of combination therapies in glioblastoma and other cancers and may significantly alter standard of care strategies for these diseases.
Collapse
Affiliation(s)
- Edwin Chang
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Chirag B. Patel
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
- Division of Neuro-Oncology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Christoph Pohling
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Caroline Young
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Jonathan Song
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Thomas Anthony Flores
- Department of Applied Physics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Yitian Zeng
- Department of Materials Science & Engineering, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Lydia-Marie Joubert
- Electron Microscopy Unit, Stellenbosch University, Stellenbosch, South Africa
| | - Hamed Arami
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Robert Sinclair
- Department of Materials Science & Engineering, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Sanjiv S. Gambhir
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
- Department of Materials Science & Engineering, Stanford University School of Medicine, Stanford, CA 94305 USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305 USA
| |
Collapse
|
604
|
Feldheim J, Kessler AF, Schmitt D, Wilczek L, Linsenmann T, Dahlmann M, Monoranu CM, Ernestus RI, Hagemann C, Löhr M. Expression of activating transcription factor 5 (ATF5) is increased in astrocytomas of different WHO grades and correlates with survival of glioblastoma patients. Onco Targets Ther 2018; 11:8673-8684. [PMID: 30584325 PMCID: PMC6287669 DOI: 10.2147/ott.s176549] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background ATF5 suppresses differentiation of neuroprogenitor cells and is overexpressed in glioblastoma (GBM). A reduction of its expression leads to apoptotic GBM cell death. Data on ATF5 expression in astrocytoma WHO grade II (low-grade astrocytoma [LGA]) are scarce and lacking on recurrent GBM. Patients and methods ATF5 mRNA was extracted from frozen samples of patients’ GBM (n=79), LGA (n=40), and normal brain (NB, n=10), quantified by duplex qPCR and correlated with retrospectively collected clinical data. ATF5 protein expression was evaluated by measuring staining intensity on immunohistochemistry. Results ATF5 mRNA was overexpressed in LGA (sevenfold, P<0.001) and GBM (tenfold, P<0.001) compared to NB, which was confirmed on protein level. Although ATF5 mRNA expression in GBM showed a considerable fluctuation range, groups of varying biological behavior, that is, local/multifocal growth or primary tumor/relapse and the tumor localization at diagnosis, were not significantly different. ATF5 mRNA correlated with the patients’ age (r=0.339, P=0.028) and inversely with Ki67-staining (r=−0.421, P=0.007). GBM patients were allocated to a low and a high ATF5 expression group by the median ATF5 overexpression compared to NB. Kaplan–Meier analysis and Cox regression indicated that ATF5 mRNA expression significantly correlated with short-term survival (t,12 months, median survival 18 vs 13 months, P=0.022, HR 2.827) and progression-free survival (PFS) (12 vs 6 months, P=0.024). This advantage vanished after 24 months (P=0.084). Conclusion ATF5 mRNA expression could be identified as an additional, though not independent factor correlating with overall survival and PFS. Since its inhibition might lead to the selective death of glioma cells, it might serve as a potential ubiquitous therapeutic target in astrocytic tumors.
Collapse
Affiliation(s)
- Jonas Feldheim
- Department of Neurosurgery, Tumorbiology Laboratory, University of Würzburg, Würzburg, Germany,
| | - Almuth F Kessler
- Department of Neurosurgery, Tumorbiology Laboratory, University of Würzburg, Würzburg, Germany,
| | - Dominik Schmitt
- Department of Neurosurgery, Tumorbiology Laboratory, University of Würzburg, Würzburg, Germany,
| | - Lara Wilczek
- Department of Neurosurgery, Tumorbiology Laboratory, University of Würzburg, Würzburg, Germany,
| | - Thomas Linsenmann
- Department of Neurosurgery, Tumorbiology Laboratory, University of Würzburg, Würzburg, Germany,
| | - Mathias Dahlmann
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Camelia M Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, Tumorbiology Laboratory, University of Würzburg, Würzburg, Germany,
| | - Carsten Hagemann
- Department of Neurosurgery, Tumorbiology Laboratory, University of Würzburg, Würzburg, Germany,
| | - Mario Löhr
- Department of Neurosurgery, Tumorbiology Laboratory, University of Würzburg, Würzburg, Germany,
| |
Collapse
|
605
|
Fatehi M, Hunt C, Ma R, Toyota BD. Persistent Disparities in Survival for Patients with Glioblastoma. World Neurosurg 2018; 120:e511-e516. [DOI: 10.1016/j.wneu.2018.08.114] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 08/16/2018] [Indexed: 11/26/2022]
|
606
|
Jhaveri J, Cheng E, Tian S, Buchwald Z, Chowdhary M, Liu Y, Gillespie TW, Olson JJ, Diaz AZ, Voloschin A, Eaton BR, Crocker IR, McDonald MW, Curran WJ, Patel KR. Proton vs. Photon Radiation Therapy for Primary Gliomas: An Analysis of the National Cancer Data Base. Front Oncol 2018; 8:440. [PMID: 30547008 PMCID: PMC6279888 DOI: 10.3389/fonc.2018.00440] [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: 07/18/2018] [Accepted: 09/20/2018] [Indexed: 11/13/2022] Open
Abstract
Background: To investigate the impact of proton radiotherapy (PBT) on overall survival (OS) and evaluate PBT usage trends for patients with gliomas in the National Cancer Data Base (NCDB). Methods: Patients with a diagnosis of World Health Organization (WHO) Grade I-IV glioma treated with definitive radiation therapy (RT) between the years of 2004–13 were identified. Patients were stratified based on WHO Grade and photon radiotherapy (XRT) vs. PBT. Univariate (UVA) and multivariable analysis (MVA) with OS were performed by Cox proportional hazards model and log-rank tests. Propensity score (PS) weighting was utilized to account for differences in patient characteristics and to minimize selection bias. Results: There were a total of 49,405 patients treated with XRT and 170 patients treated with PBT. Median follow-up time was 62.1 months. On MVA, the following factors were associated with receipt of PBT (all p < 0.05): WHO Grade I-II gliomas, treatment at an academic/research program, west geographic facility location, and surgical resection. After PS weighting, all patients treated with PBT were found to have superior median and 5 year survival than patients treated with XRT: 45.9 vs. 29.7 months (p = 0.009) and 46.1 vs. 35.5% (p = 0.0160), respectively. Conclusions: PBT is associated with improved OS compared to XRT for patients with gliomas. This finding warrants verification in the randomized trial setting in order to account for potential patient imbalances not adequately captured by the NCDB, such as tumor molecular characteristics and patient performance status. Importance of the Study: This is the first study that compares the outcomes of patients treated with photon based radiotherapy vs. proton based radiotherapy for patients with gliomas. In this retrospective analysis, the results demonstrate that proton therapy is associated with improved outcomes which support ongoing prospective, randomized clinical trials comparing the two modalities in patients with gliomas.
Collapse
Affiliation(s)
- Jaymin Jhaveri
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - En Cheng
- Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Sibo Tian
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Zachary Buchwald
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Mudit Chowdhary
- Department of Radiation Oncology, Rush University, Chicago, IL, United States
| | - Yuan Liu
- Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | | | - Jeffrey J Olson
- Department of Neurosurgery and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Aidnag Z Diaz
- Department of Radiation Oncology, Rush University, Chicago, IL, United States
| | - Alfredo Voloschin
- Department of Hematology and Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Bree R Eaton
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Ian R Crocker
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Mark W McDonald
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Walter J Curran
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Kirtesh R Patel
- Department of Therapeutic Radiology, Yale University, New Haven, CT, United States
| |
Collapse
|
607
|
Branter J, Basu S, Smith S. Tumour treating fields in a combinational therapeutic approach. Oncotarget 2018; 9:36631-36644. [PMID: 30564303 PMCID: PMC6290966 DOI: 10.18632/oncotarget.26344] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 10/24/2018] [Indexed: 12/15/2022] Open
Abstract
The standard of care for patients with newly diagnosed Glioblastoma multiforme (GBM) has remained unchanged since 2005, with patients undergoing maximal surgical resection, followed by radiotherapy plus concomitant and maintenance Temozolomide. More recently, Tumour treating fields (TTFields) therapy has become FDA approved for adult recurrent and adult newly-diagnosed GBM following the EF-11 and EF-14 trials, respectively. TTFields is a non-invasive anticancer treatment which utilizes medium frequency alternating electric fields to target actively dividing cancerous cells. TTFields selectively targets cells within mitosis through interacting with key mitotic proteins to cause mitotic arrest and cell death. TTFields therapy presents itself as a candidate for the combinational therapy route due to the lack of overlapping toxicities associated with electric fields. Here we review current literature pertaining to TTFields in combination with alkylating agents, radiation, anti-angiogenics, mitotic inhibitors, immunotherapies, and also with novel agents. This review highlights the observed synergistic and additive effects of combining TTFields with various other therapies, as well highlighting the strategies relating to combinations with electric fields.
Collapse
Affiliation(s)
- Joshua Branter
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Surajit Basu
- Queen's Medical Centre, Department of Neurosurgery, Nottingham, UK
| | - Stuart Smith
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
608
|
Abstract
The physical characteristics of proton therapy result in steeper dose gradients and superior dose conformality compared to photon therapy. These properties render proton therapy ideal for skull base tumors requiring dose escalation for optimal tumor control, and may also be beneficial for brain tumors as a means of mitigating radiation-related adverse effects. This review summarizes the literature regarding the role of proton therapy compared to photon therapy in the treatment of adult brain and skull base tumors.
Collapse
Affiliation(s)
- Safia K Ahmed
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN.
| | - Robert L Foote
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| |
Collapse
|
609
|
Doyle SP, Gurbani SS, Ross AS, Rosen H, Barrett CD, Olson JJ, Shim H, Shu HK, Sengupta S. The role of erlotinib and the Optune device in a patient with an epidermal growth factor receptor viii amplified glioblastoma. Oxf Med Case Reports 2018; 2018:omy095. [PMID: 30410775 PMCID: PMC6217712 DOI: 10.1093/omcr/omy095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/17/2018] [Accepted: 08/27/2018] [Indexed: 12/03/2022] Open
Abstract
The standard treatment for patients diagnosed with glioblastoma is surgical resection of tumor followed by high dose radiation and chemotherapy with temozolomide. For patients who experience allergic reactions to temozolomide despite desensitization protocols, alternative therapies must be considered. In this report, we present such a patient who then received treatment with an epidermal growth factor receptor inhibitor, erlotinib, concurrent with a tumor-treating field device, Optune. Through this combination of a targeted molecular therapy and the Optune device, the patient has been able to achieve stable disease 9 months after completing radiation.
Collapse
Affiliation(s)
- Sean P Doyle
- Emory University School of Medicine, Atlanta, GA, USA
| | | | - Alexandra S Ross
- Departments of Neurology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Havi Rosen
- Departments of Neurology and Medical Oncology, Emory University, Atlanta, GA, USA
| | | | - Jeffrey J Olson
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA
| | - Hui-Kuo Shu
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA
| | - Soma Sengupta
- Departments of Neurology and Medical Oncology, Emory University, Atlanta, GA, USA
| |
Collapse
|
610
|
Abstract
OBJECTIVE To introduce effectiveness of tumor treating fields (TTFields), how to care for the patient with this type of treatment, and the critical role the nurse plays in educating the patient about this innovative treatment. DATA SOURCES Published research and articles in both nursing and medical journals. CONCLUSION TTFields are an antimitotic therapy delivered via transducer arrays that are worn on the scalp to treat newly diagnosed and recurrent glioblastoma, the most aggressive primary brain cancer. Oncology nurses are integral in educating and supporting the patient in its use and managing its of treatment. IMPLICATIONS FOR NURSING PRACTICE Nurses are on the front line of educating the patient, caregivers, and the larger body of clinicians who deliver care to these patients. Education provided by nurses increases the patients' knowledge, and thus compliance, as well as the overall outcome through proper usage of TTFields.
Collapse
|
611
|
Haupt M. The Study Was Subject to Considerable Bias. DEUTSCHES ARZTEBLATT INTERNATIONAL 2018; 115:674. [PMID: 30381134 PMCID: PMC6234465 DOI: 10.3238/arztebl.2018.0674a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
|
612
|
Abstract
OBJECTIVE To describe the indications for convection-enhanced delivery in the treatment of glioblastoma, highlighting candidates for the delivery method, mechanics of drug delivery, and management of acute and long-term complications. DATA SOURCES A conceptual framework drawn from published literature as well as author's expert experiences. CONCLUSION Convection-enhanced delivery is an established method of delivering new therapies to patients with glioblastoma. Management of both acute and long-term complications is often drug dependent. IMPLICATIONS FOR NURSING PRACTICE Nurses should be able to recognize and manage potential complications during the infusion of agents delivered via convection-enhanced delivery. Post-infusion symptoms may worsen because of immunologic responses related to the drug and management should be directed toward symptom relief and support without interference on the immunologic response.
Collapse
|
613
|
Regulation of human glioma cell migration, tumor growth, and stemness gene expression using a Lck targeted inhibitor. Oncogene 2018; 38:1734-1750. [PMID: 30353164 PMCID: PMC6462869 DOI: 10.1038/s41388-018-0546-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 10/03/2018] [Accepted: 10/06/2018] [Indexed: 11/28/2022]
Abstract
Migration of human glioma cells (hGCs) within the brain parenchyma makes glioblastoma one of the most aggressive and lethal tumors. Studies of the cellular and molecular mechanisms underlying hGC migration are hindered by the limitations of existing glioma models. Here we developed a dorsal root ganglion axon-oligodendrocyte-hGC co-culture to study in real time the migration and interaction of hGCs with their microenvironment. hGCs interact with myelinated and non-myelinated axons through the formation of pseudopodia. Isolation of pseudopodia-localized polysome-bound RNA reveals transcripts of Lck, Paxillin, Crk-II, and Rac1 that undergo local translation. Inhibition of Lck phosphorylation using a small-molecule inhibitor (Lck-I), blocks the phosphorylation of Paxillin and Crk-II, the formation of pseudopodia and the migration of hGCs. In vivo intraventricular administration of the Lck-I using an orthotopic xenograft glioma model, results in statistically significant inhibition of tumor size and significant down-regulation of Nanog-targeted genes, which are associated with glioblastoma patient survival. Moreover, treatment of human glioma stem cells (hGSCs) with Lck-I, results in significant inhibition of self-renewal and tumor-sphere formation. The involvement of Lck in different levels of glioma malignant progression, such as migration, tumor growth, and regulation of cancer stemness, makes Lck a potentially important therapeutic target for human glioblastomas.
Collapse
|
614
|
Kaley T, Touat M, Subbiah V, Hollebecque A, Rodon J, Lockhart AC, Keedy V, Bielle F, Hofheinz RD, Joly F, Blay JY, Chau I, Puzanov I, Raje NS, Wolf J, DeAngelis LM, Makrutzki M, Riehl T, Pitcher B, Baselga J, Hyman DM. BRAF Inhibition in BRAFV600-Mutant Gliomas: Results From the VE-BASKET Study. J Clin Oncol 2018; 36:3477-3484. [PMID: 30351999 PMCID: PMC6286161 DOI: 10.1200/jco.2018.78.9990] [Citation(s) in RCA: 242] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Purpose BRAFV600 mutations are frequently found in several glioma subtypes, including pleomorphic xanthoastrocytoma (PXA) and ganglioglioma and much less commonly in glioblastoma. We sought to determine the activity of vemurafenib, a selective inhibitor of BRAFV600, in patients with gliomas that harbor this mutation. Patients and Methods The VE-BASKET study was an open-label, nonrandomized, multicohort study for BRAFV600-mutant nonmelanoma cancers. Patients with BRAFV600-mutant glioma received vemurafenib 960 mg twice per day continuously until disease progression, withdrawal, or intolerable adverse effects. Key end points included confirmed objective response rate by RECIST version 1.1, progression-free survival, overall survival, and safety. Results Twenty-four patients (median age, 32 years; 18 female and six male patients) with glioma, including malignant diffuse glioma (n = 11; six glioblastoma and five anaplastic astrocytoma), PXA (n = 7), anaplastic ganglioglioma (n = 3), pilocytic astrocytoma (n = 2), and high-grade glioma, not otherwise specified (n = 1), were treated. Confirmed objective response rate was 25% (95% CI, 10% to 47%) and median progression-free survival was 5.5 months (95% CI, 3.7 to 9.6 months). In malignant diffuse glioma, best response included one partial response and five patients with stable disease, two of whom had disease stabilization that lasted more than 1 year. In PXA, best response included one complete response, two partial responses, and three patients with stable disease. Additional partial responses were observed in patients with pilocytic astrocytoma and anaplastic ganglioglioma (one each). The safety profile of vemurafenib was generally consistent with that of previously published studies. Conclusion Vemurafenib demonstrated evidence of durable antitumor activity in some patients with BRAFV600-mutant gliomas, although efficacy seemed to vary qualitatively by histologic subtype. Additional study is needed to determine the optimal use of vemurafenib in patients with primary brain tumors and to identify the mechanisms driving differential responses across histologic subsets.
Collapse
Affiliation(s)
- Thomas Kaley
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Mehdi Touat
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Vivek Subbiah
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Antoine Hollebecque
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Jordi Rodon
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - A Craig Lockhart
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Vicki Keedy
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Franck Bielle
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Ralf-Dieter Hofheinz
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Florence Joly
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Jean-Yves Blay
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Ian Chau
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Igor Puzanov
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Noopur S Raje
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Jurgen Wolf
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Lisa M DeAngelis
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Martina Makrutzki
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Todd Riehl
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Bethany Pitcher
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - Jose Baselga
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| | - David M Hyman
- Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Memorial Sloan Kettering Cancer Center; Thomas Kaley, Lisa M. DeAngelis, Jose Baselga, and David M. Hyman, Weill Cornell Medical College, New York; Igor Puzanov, Roswell Park Cancer Institute, Buffalo, NY; Mehdi Touat and Antoine Hollebecque, Institut Gustave Roussy, Villejuif; Franck Bielle, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Paris; Florence Joly, Centre François Baclesse, Caen; Jean-Yves Blay, Centre Léon Bérard, Lyon, France; Vivek Subbiah, MD Anderson Cancer Center, Houston, TX; Jordi Rodon, Vall d'Hebron University Hospital, Barcelona, Spain; A. Craig Lockhart, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL; Vicki Keedy, Vanderbilt University Medical Center, Nashville, TN; Ralf-Dieter Hofheinz, Universitätsmedizin Mannheim, Mannheim; Jurgen Wolf, Universitätsklinikum Köln, Cologne, Germany; Ian Chau, The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Noopur S. Raje, Massachusetts General Hospital, Boston, MA; Martina Makrutzki, F Hoffmann-La Roche, Basel, Switzerland; Todd Riehl, Genentech, South San Francisco, CA; and Bethany Pitcher, F Hoffmann-La Roche, Mississauga, Ontario, Canada
| |
Collapse
|
615
|
Proliferation arrest, selectivity, and chemosensitivity enhancement of cancer cells treated by a low-intensity alternating electric field. Biomed Microdevices 2018; 20:90. [DOI: 10.1007/s10544-018-0339-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
616
|
Shteingauz A, Porat Y, Voloshin T, Schneiderman RS, Munster M, Zeevi E, Kaynan N, Gotlib K, Giladi M, Kirson ED, Weinberg U, Kinzel A, Palti Y. AMPK-dependent autophagy upregulation serves as a survival mechanism in response to Tumor Treating Fields (TTFields). Cell Death Dis 2018; 9:1074. [PMID: 30341282 PMCID: PMC6195570 DOI: 10.1038/s41419-018-1085-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 01/04/2023]
Abstract
Tumor Treating Fields (TTFields), an approved treatment modality for glioblastoma, are delivered via non-invasive application of low-intensity, intermediate-frequency, alternating electric fields. TTFields application leads to abnormal mitosis, aneuploidy, and increased cell granularity, which are often associated with enhancement of autophagy. In this work, we evaluated whether TTFields effected the regulation of autophagy in glioma cells. We found that autophagy is upregulated in glioma cells treated with TTFields as demonstrated by immunoblot analysis of the lipidated microtubule-associated protein light chain 3 (LC3-II). Fluorescence and transmission electron microscopy demonstrated the presence of LC3 puncta and typical autophagosome-like structures in TTFields-treated cells. Utilizing time-lapse microscopy, we found that the significant increase in the formation of LC3 puncta was specific to cells that divided during TTFields application. Evaluation of selected cell stress parameters revealed an increase in the expression of the endoplasmic reticulum (ER) stress marker GRP78 and decreased intracellular ATP levels, both of which are indicative of increased proteotoxic stress. Pathway analysis demonstrated that TTFields-induced upregulation of autophagy is dependent on AMP-activated protein kinase (AMPK) activation. Depletion of AMPK or autophagy-related protein 7 (ATG7) inhibited the upregulation of autophagy in response to TTFields, as well as sensitized cells to the treatment, suggesting that cancer cells utilize autophagy as a resistance mechanism to TTFields. Combining TTFields with the autophagy inhibitor chloroquine (CQ) resulted in a significant dose-dependent reduction in cell growth compared with either TTFields or CQ alone. These results suggest that dividing cells upregulate autophagy in response to aneuploidy and ER stress induced by TTFields, and that AMPK serves as a key regulator of this process.
Collapse
|
617
|
Akay M, Hite J, Avci NG, Fan Y, Akay Y, Lu G, Zhu JJ. Drug Screening of Human GBM Spheroids in Brain Cancer Chip. Sci Rep 2018; 8:15423. [PMID: 30337660 PMCID: PMC6194126 DOI: 10.1038/s41598-018-33641-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), an extremely invasive and high-grade (grade IV) glioma, is the most common and aggressive form of brain cancer. It has a poor prognosis, with a median overall survival of only 11 months in the general GBM population and 14.6 to 21 months in clinical trial participants with standard GBM therapies, including maximum safe craniotomy, adjuvant radiation, and chemotherapies. Therefore, new approaches for developing effective treatments, such as a tool for assessing tumor cell drug response before drug treatments are administered, are urgently needed to improve patient survival. To address this issue, we developed an improved brain cancer chip with a diffusion prevention mechanism that blocks drugs crossing from one channel to another. In the current study, we demonstrate that the chip has the ability to culture 3D spheroids from patient tumor specimen-derived GBM cells obtained from three GBM patients. Two clinical drugs used to treat GBM, temozolomide (TMZ) and bevacizumab (Avastin, BEV), were applied and a range of relative concentrations was generated by the microfluidic channels in the brain cancer chip. The results showed that TMZ works more effectively when used in combination with BEV compared to TMZ alone. We believe that this low-cost brain cancer chip could be further developed to generate optimal combination of chemotherapy drugs tailored to individual GBM patients.
Collapse
Affiliation(s)
- Metin Akay
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA.
| | - John Hite
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Naze Gul Avci
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Yantao Fan
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Yasemin Akay
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Guangrong Lu
- Mischer Neuroscience Associates and the Vivian L. Smith Department of Neurosurgery University of Texas Health Science Center in Houston, UTHealth and Memorial Hermann, 6400 Fannin St. Suite 2800, Houston, TX, 77030, USA
| | - Jay-Jiguang Zhu
- Mischer Neuroscience Associates and the Vivian L. Smith Department of Neurosurgery University of Texas Health Science Center in Houston, UTHealth and Memorial Hermann, 6400 Fannin St. Suite 2800, Houston, TX, 77030, USA
| |
Collapse
|
618
|
Rahman M, Dastmalchi F, Karachi A, Mitchell D. The role of CMV in glioblastoma and implications for immunotherapeutic strategies. Oncoimmunology 2018; 8:e1514921. [PMID: 30546954 PMCID: PMC6287786 DOI: 10.1080/2162402x.2018.1514921] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/09/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022] Open
Abstract
Controversy surrounds the role of cytomegalovirus (CMV) in glioblastoma (GBM). However, several studies have shown that CMV nucleic acids and proteins are present within GBM tumor tissue. CMV has been implicated in GBM pathogenesis by affecting tumor stem cell factors, angiogenesis and immune pathways. Anti-viral therapy has not been found to definitively improve outcomes for patients with GBM. Several studies have leveraged CMV by targeting CMV antigens using ex-vivo expanded T cells or dendritic cell vaccines. The initial results from these studies are promising and larger studies are underway.
Collapse
Affiliation(s)
- Maryam Rahman
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Farhad Dastmalchi
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Aida Karachi
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Duane Mitchell
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
619
|
Robins HI, Nguyen HN, Field A, Howard S, Salamat S, Deming DA. Molecular Evolution of a Glioblastoma Controlled With Tumor Treating Fields and Concomitant Temozolomide. Front Oncol 2018; 8:451. [PMID: 30374424 PMCID: PMC6196276 DOI: 10.3389/fonc.2018.00451] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/26/2018] [Indexed: 12/02/2022] Open
Abstract
Tumor Treating Field (TTFields) therapy has demonstrated efficacy in a Phase 3 study of newly diagnosed glioblastoma (GB) following radiation (RT) and temozolomide (TMZ). We report the appearance of an isolated satellite anterior temporal lobe lesion, 2 months post primary RT/TMZ directed at the primary GB (MGMT methylated) parietal lobe lesion and one adjuvant cycle of TMZ and TTFields. The mean RT dose delivered to the temporal lobe lesion was negligible, i.e., 4.53 ± 0.95 Gy. Mapping of the generated TTFields demonstrated that both lesions were encompassed by a field intensity in a therapeutic range. The temporal lobe lesion remained under the control of TTFields up to 12 months, at which point progression on a T1 contrast MRI resulted in surgery and a definitive diagnosis of GB without MGMT methylation. The primary parietal lobe at this time was in remission. Molecular sequencing on the GB tissue from multiple time points demonstrates clonal evolution of the cancer over time and in response to treatment.
Collapse
Affiliation(s)
- H Ian Robins
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - HuyTram N Nguyen
- University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Aaron Field
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Steven Howard
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Shahriar Salamat
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Dustin A Deming
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
620
|
Hoang-Minh LB, Siebzehnrubl FA, Yang C, Suzuki-Hatano S, Dajac K, Loche T, Andrews N, Schmoll Massari M, Patel J, Amin K, Vuong A, Jimenez-Pascual A, Kubilis P, Garrett TJ, Moneypenny C, Pacak CA, Huang J, Sayour EJ, Mitchell DA, Sarkisian MR, Reynolds BA, Deleyrolle LP. Infiltrative and drug-resistant slow-cycling cells support metabolic heterogeneity in glioblastoma. EMBO J 2018; 37:embj.201798772. [PMID: 30322894 DOI: 10.15252/embj.201798772] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 08/23/2018] [Accepted: 08/24/2018] [Indexed: 01/01/2023] Open
Abstract
Metabolic reprogramming has been described in rapidly growing tumors, which are thought to mostly contain fast-cycling cells (FCCs) that have impaired mitochondrial function and rely on aerobic glycolysis. Here, we characterize the metabolic landscape of glioblastoma (GBM) and explore metabolic specificities as targetable vulnerabilities. Our studies highlight the metabolic heterogeneity in GBM, in which FCCs harness aerobic glycolysis, and slow-cycling cells (SCCs) preferentially utilize mitochondrial oxidative phosphorylation for their functions. SCCs display enhanced invasion and chemoresistance, suggesting their important role in tumor recurrence. SCCs also demonstrate increased lipid contents that are specifically metabolized under glucose-deprived conditions. Fatty acid transport in SCCs is targetable by pharmacological inhibition or genomic deletion of FABP7, both of which sensitize SCCs to metabolic stress. Furthermore, FABP7 inhibition, whether alone or in combination with glycolysis inhibition, leads to overall increased survival. Our studies reveal the existence of GBM cell subpopulations with distinct metabolic requirements and suggest that FABP7 is central to lipid metabolism in SCCs and that targeting FABP7-related metabolic pathways is a viable therapeutic strategy.
Collapse
Affiliation(s)
- Lan B Hoang-Minh
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Florian A Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, UK
| | - Changlin Yang
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Silveli Suzuki-Hatano
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Kyle Dajac
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Tyler Loche
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Nicholas Andrews
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Michael Schmoll Massari
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jaimin Patel
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Krisha Amin
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Alvin Vuong
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Ana Jimenez-Pascual
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, UK
| | - Paul Kubilis
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Craig Moneypenny
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, USA
| | - Christina A Pacak
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jianping Huang
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Elias J Sayour
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Duane A Mitchell
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Matthew R Sarkisian
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Brent A Reynolds
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA .,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Loic P Deleyrolle
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA .,Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
621
|
MSI1 associates glioblastoma radioresistance via homologous recombination repair, tumor invasion and cancer stem-like cell properties. Radiother Oncol 2018; 129:352-363. [PMID: 30322656 DOI: 10.1016/j.radonc.2018.09.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 09/17/2018] [Accepted: 09/21/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) is the most common brain malignancy in adults, and currently available GBM treatments present several unique challenges. It is known that GBM involves cancer stem-like cells (CSCs) and tumor cells that aggressively invade normal brain tissues, and both cell types may cause resistance to radiotherapy (RT) and are thus responsible for therapeutic failure. The radioresistance of GBM cells relies on the efficient activation of the DNA damage response (DDR), but the mechanisms linking this response with stem-cell status and tumor invasion remain unclear. MATERIALS AND METHODS We used irradiation to treat patient-derived GBM (Par) cells and then purified radioresistant GBM (R2M2) cells through two rounds of irradiation and an invasion assay. Musashi-1 (MSI1) is a neural stem-cell marker and key oncogenic factor of GBM. We identified MSI1 expression to predict radioresistance through silencing an MSI1-high-expressing R2M2 cell line or inducing overexpression in a Par cell line with low/no MSI1 expression and assessing the subsequent DDR. RESULT MSI1 enhances tumor invasion via VCAM1 and modulates GBM radioresistance via the hyperactivation of the DDR through increasing homologous recombination repair and evading apoptosis. MSI1 knockdown induces DNA damage accumulation in irradiated GBM cells and promotes their depletion in vitro; MSI1 knockdown also inhibits the formation of GBMs generated by irradiated xeno-transplanted cells. MSI1 inhibition may radiosensitize tumors, prevent CSC-positive selection induced by RT, and reduce tumor invasion. CONCLUSION MSI1 may involve in regulating GBM radioresistance, invasion, and recurrence and could be a novel target for GBM treatment.
Collapse
|
622
|
Buchroithner J, Erhart F, Pichler J, Widhalm G, Preusser M, Stockhammer G, Nowosielski M, Iglseder S, Freyschlag CF, Oberndorfer S, Bordihn K, von Campe G, Hoffermann M, Ruckser R, Rössler K, Spiegl-Kreinecker S, Fischer MB, Czech T, Visus C, Krumpl G, Felzmann T, Marosi C. Audencel Immunotherapy Based on Dendritic Cells Has No Effect on Overall and Progression-Free Survival in Newly Diagnosed Glioblastoma: A Phase II Randomized Trial. Cancers (Basel) 2018; 10:E372. [PMID: 30301187 PMCID: PMC6210090 DOI: 10.3390/cancers10100372] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/22/2018] [Accepted: 09/30/2018] [Indexed: 11/17/2022] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells that are capable of priming anti-tumor immune responses, thus serving as attractive tools to generate tumor vaccines. In this multicentric randomized open-label phase II study, we investigated the efficacy of vaccination with tumor lysate-charged autologous DCs (Audencel) in newly diagnosed glioblastoma multiforme (GBM). Patients aged 18 to 70 years with histologically proven primary GBM and resection of at least 70% were randomized 1:1 to standard of care (SOC) or SOC plus vaccination (weekly intranodal application in weeks seven to 10, followed by monthly intervals). The primary endpoint was progression-free survival at 12 months. Secondary endpoints were overall survival, safety, and toxicity. Seventy-six adult patients were analyzed in this study. Vaccinations were given for seven (3⁻20) months on average. No severe toxicity was attributable to vaccination. Seven patients showed flu-like symptoms, and six patients developed local skin reactions. Progression-free survival at 12 months did not differ significantly between the control and vaccine groups (28.4% versus 24.5%, p = 0.9975). Median overall survival was similar with 18.3 months (vaccine: 564 days, 95% CI: 436⁻671 versus control: 568 days, 95% CI: 349⁻680; p = 0.89, harzard ratio (HR) 0.99). Hence, in this trial, the clinical outcomes of patients with primary GBM could not be improved by the addition of Audencel to SOC.
Collapse
Affiliation(s)
- Johanna Buchroithner
- University Clinic for Neurosurgery, Kepler University Hospital, Johannes Kepler University, Wagner-Jauregg-Weg 15, 4020 Linz, Austria.
| | - Friedrich Erhart
- Department of Neurosurgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
- Institute of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Josef Pichler
- Department of Internal Medicine and Neurooncology, Kepler University Hospital, Johannes Kepler University, Wagner-Jauregg-Weg 15, 4020 Linz, Austria.
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Matthias Preusser
- Clinical Division of Medical Oncology, Department for Internal Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Günther Stockhammer
- Department of Neurology, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria.
| | - Martha Nowosielski
- Department of Neurology, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria.
| | - Sarah Iglseder
- Department of Neurology, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria.
| | - Christian F Freyschlag
- Department of Neurosurgery, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria.
| | - Stefan Oberndorfer
- Department of Neurology, University Clinic St. Pölten, Karl Landsteiner Privat Universität, Dunant-Platz 1, 3100 St. Pölten, Austria.
| | - Karin Bordihn
- Department of Neurosurgery, Landeskrankenhaus Salzburg, University Clinic of the Paracelsus Private Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria.
| | - Gord von Campe
- Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 29, 8036 Graz, Austria.
| | - Markus Hoffermann
- Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 29, 8036 Graz, Austria.
| | - Reinhard Ruckser
- Department of Internal Medicine 2, Donauspital, Langobardenstraße 122, 1220 Vienna, Austria.
| | - Karl Rössler
- Department of Neurosurgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany.
| | - Sabine Spiegl-Kreinecker
- University Clinic for Neurosurgery, Kepler University Hospital, Johannes Kepler University, Wagner-Jauregg-Weg 15, 4020 Linz, Austria.
| | - Michael B Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Thomas Czech
- Department of Neurosurgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Carmen Visus
- Activartis Biotech GmbH, Wilhelminenstraße 91/IIf, 1160 Vienna, Austria.
| | - Günther Krumpl
- Activartis Biotech GmbH, Wilhelminenstraße 91/IIf, 1160 Vienna, Austria.
| | - Thomas Felzmann
- Activartis Biotech GmbH, Wilhelminenstraße 91/IIf, 1160 Vienna, Austria.
| | - Christine Marosi
- Clinical Division of Medical Oncology, Department for Internal Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
623
|
Selective toxicity of tumor treating fields to melanoma: an in vitro and in vivo study. Cell Death Discov 2018; 4:46. [PMID: 30302280 PMCID: PMC6170382 DOI: 10.1038/s41420-018-0106-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/07/2018] [Accepted: 07/10/2018] [Indexed: 11/08/2022] Open
Abstract
Tumor treating fields (TTFs) are a newly developed cancer therapy technology using an alternating electric field that may be a possible candidate for overcoming the limitations of conventional treatment methods currently used in cancer treatment. Although clinical results using TTFs appear promising, concerns regarding side effects must be clarified to demonstrate the effectiveness of this treatment method. To investigate the side effects of TTF treatment, the damage to normal cell lines and normal tissue of a mouse model was compared with the damage to tumor cells and tumors in a mouse model after TTF treatment. No serious damage was found in the normal cells and normal tissues of the mouse model, suggesting that the side effects of TTF treatment may not be serious. Our evidence based on in vitro and in vivo experiments suggests that TTF may cause selective damage to cancer cells, further demonstrating the potential of TTF as an attractive alternative to conventional cancer treatment modalities.
Collapse
|
624
|
Navone SE, Guarnaccia L, Locatelli M, Rampini P, Caroli M, La Verde N, Gaudino C, Bettinardi N, Riboni L, Marfia G, Campanella R. Significance and Prognostic Value of The Coagulation Profile in Patients with Glioblastoma: Implications for Personalized Therapy. World Neurosurg 2018; 121:e621-e629. [PMID: 30292037 DOI: 10.1016/j.wneu.2018.09.177] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Coagulation is an important aspect of the vascular microenvironment in which brain tumors evolve. Patients with tumor often show aberrant coagulation and fibrinolysis activation. In particular, glioblastoma (GBM), the most aggressive primary brain tumor, is associated with a state of hypercoagulability, and venous thromboembolism is a common complication of this cancer and its treatment. Our study aims to investigate the clinical and prognostic significance of routine laboratory tests to assess the coagulative state of patients with brain tumors, to identify potential new prognostic factors and targets for personalized therapy. METHODS Blood samples were collected from patients with GBM (n = 58) and patients with meningioma (MNG, n = 22), before any treatment. The parameters analyzed were prothrombin time (PT), activated partial thromboplastin time (aPTT), D dimer (DD), fibrinogen, von Willebrand factor (VWF), leukocyte count, and hemoglobin levels. RESULTS Plasma levels of PT and aPTT were significantly reduced in GBMs compared with MNGs (P < 0.05), whereas DD, VWF:Ag levels, and leukocyte count were significantly higher in GBMs than in MNGs (P < 0.01). Furthermore, we observed that patients with GBM with reduced PT and aPTT and high levels of DD and VWF, defined as hypercoagulable patients, showed reduced overall survival (P < 0.05) compared with nonhypercoagulable patients. CONCLUSIONS Our data support the assumption that patients with GBM show a plasma hypercoagulable profile and that coagulation profile is related to adverse outcome in patients with GBM. If confirmed, hypercoagulability could play an important role as a prognostic factor of the disease and in the decision of an antithrombotic prophylaxis.
Collapse
Affiliation(s)
- Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, University of Milan, Milan, Italy
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, University of Milan, Milan, Italy
| | - Marco Locatelli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, University of Milan, Milan, Italy
| | - Paolo Rampini
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, University of Milan, Milan, Italy
| | - Manuela Caroli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, University of Milan, Milan, Italy
| | - Nicla La Verde
- Oncology Unit, Fatebenefratelli and Oftalmico Hospital, Milan, Italy
| | - Chiara Gaudino
- Department of Neuroradiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Nora Bettinardi
- Central Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, Milan, Italy
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, University of Milan, Milan, Italy.
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, University of Milan, Milan, Italy
| |
Collapse
|
625
|
Li J, Zhu Y, Wang H, Ji X. Targeting Long Noncoding RNA in Glioma: A Pathway Perspective. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:431-441. [PMID: 30388617 PMCID: PMC6202792 DOI: 10.1016/j.omtn.2018.09.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/15/2018] [Accepted: 09/26/2018] [Indexed: 02/09/2023]
Abstract
Long noncoding RNAs (lncRNAs) participate extensively in biological processes of various cancers. The majority of these transcripts are uniquely expressed in differentiated tissues or specific cancer types. lncRNAs are aberrantly expressed in gliomas and exert diverse functions. In this article, we provided an overview of how lncRNAs regulate cellular processes in glioma, enumerated the lncRNAs that may act as glioma biomarkers, and showed their potential clinical implications.
Collapse
Affiliation(s)
- Junyang Li
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Yihao Zhu
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China.
| | - Xiangjun Ji
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
626
|
Repka MC, Lei S, Campbell L, Suy S, Voyadzis JM, Kalhorn C, McGrail K, Jean W, Subramaniam DS, Lischalk JW, Collins SP, Collins BT. Long-Term Outcomes Following Conventionally Fractionated Stereotactic Boost for High-Grade Gliomas in Close Proximity to Critical Organs at Risk. Front Oncol 2018; 8:373. [PMID: 30254985 PMCID: PMC6141832 DOI: 10.3389/fonc.2018.00373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/21/2018] [Indexed: 11/18/2022] Open
Abstract
Purpose/Objective: High-grade glioma is the most common primary malignant tumor of the CNS, with death often resulting from uncontrollable intracranial disease. Radiation dose may be limited by the tolerance of critical structures, such as the brainstem and optic apparatus. In this report, long-term outcomes in patients treated with conventionally fractionated stereotactic boost for tumors in close proximity to critical structures are presented. Materials/Methods: Patients eligible for inclusion in this single institution retrospective review had a pathologically confirmed high-grade glioma status post-surgical resection. Inclusion criteria required tumor location within one centimeter of a critical structure, including the optic chiasm, optic nerve, and brainstem. Radiation therapy consisted of external beam radiation followed by a conventionally fractionated stereotactic boost. Oncologic outcomes and toxicity were assessed. Results: Thirty patients eligible for study inclusion underwent resection of a high-grade glioma. The median initial adjuvant EBRT dose was 50 Gy with a median conventionally fractionated stereotactic boost of 10 Gy. All stereotactic treatments were given in 2 Gy daily fractions. Median follow-up time for the entire cohort was 38 months with a median overall survival of 45 months and 5-year overall survival of 32.5%. The median freedom from local progression was 45 months, and the 5-year freedom from local progression was 29.7%. Two cases of radiation retinopathy were identified following treatment. No patient experienced toxicity attributable to the optic chiasm, optic nerve, or brainstem and no grade 3+ radionecrosis was observed. Conclusions: Oncologic and toxicity outcomes in high-grade glioma patients with tumors in unfavorable locations treated with conventionally fractionated stereotactic boost are comparable to those reported in the literature. This treatment strategy is appropriate for those patients with resected high-grade glioma in close proximity to critical structures.
Collapse
Affiliation(s)
- Michael C Repka
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Siyuan Lei
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Lloyd Campbell
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Simeng Suy
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Jean-Marc Voyadzis
- Department of Neurosurgery, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Christopher Kalhorn
- Department of Neurosurgery, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Kevin McGrail
- Department of Neurosurgery, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Walter Jean
- Department of Neurological Surgery, George Washington University Hospital, Washington, DC, United States
| | - Deepa S Subramaniam
- Division of Hematology and Oncology, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Jonathan W Lischalk
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Sean P Collins
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Brian T Collins
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| |
Collapse
|
627
|
Bowden SG, Han SJ. The Evolving Role of the Oncologic Neurosurgeon: Looking Beyond Extent of Resection in the Modern Era. Front Oncol 2018; 8:406. [PMID: 30319971 PMCID: PMC6167541 DOI: 10.3389/fonc.2018.00406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/06/2018] [Indexed: 11/13/2022] Open
Abstract
Neurosurgeons have played an essential role in glioma management and research for over a century. While the past twenty years have played witness to many exciting developments in glioma biology, diagnosis, and classification, relatively few novel, effective treatment strategies have been introduced. The role of neurosurgery in glioma management has been clarified, with a large body of evidence in support of maximal safe resection. However, neurosurgeons have also played a critical role in translational research during this period. The development of new MRI technologies has benefited greatly from validation with stereotactically-targeted human tissue. Careful banking of surgically acquired tissue was key to the development of a new classification scheme for glioma. Similarly, we have garnered a considerably deeper understanding of molecular and genetic properties of glioma through analysis of large surgical specimens. As our classification schemes become more sophisticated, incorporating targeted tissue sampling into the development of novel treatment strategies becomes essential. Such ex vivo analysis could be instrumental in determining mechanisms of treatment failure or success. Modern tumor neurosurgeons should consider themselves surgical neuro-oncologists, with engagement in translational research essential to furthering the field and improving outlooks for our patients.
Collapse
Affiliation(s)
- Stephen G Bowden
- Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | - Seunggu Jude Han
- Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
628
|
Nguyen HT, Nguyen N, Liu LY, Dovek L, Lenchner D, Harris R, Ozer B, Ravelo A, Sommer N, Sim MS, Elashoff R, Green R, Nghiemphu PL, Cloughesy TF, Ellingson B, Lai A. Bevacizumab at first recurrence after standard radio-chemotherapy is associated with improved overall survival in glioblastoma patients with large tumor burden. Neurooncol Pract 2018; 6:103-111. [PMID: 31386050 DOI: 10.1093/nop/npy021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Since its approval for use in recurrent glioblastoma (GBM), the survival benefit of bevacizumab (Bev) remains to be demonstrated. To address this issue, we retrospectively examined survival from first recurrence in patients treated with Bev, lomustine (CCNU), or Bev/CCNU. Methods We identified 168 primary GBM patients diagnosed at UCLA and Kaiser Permanente LA who received upfront radio-chemotherapy, followed by Bev and/or CCNU at first recurrence. Three patient groups, contemporaneously diagnosed from 2009 through 2015, were identified: (1) patients treated with Bev alone (n = 49), (2) CCNU alone (CCNU 09-15) (n = 36), and (3) Bev/CCNU (n = 53). Another CCNU control group (n = 30) diagnosed from 2001 through 2004 (CCNU 01-04) was also derived. We measured tumor size at first recurrence treatment initiation, using bidimensional (2D) and volumetric (3D) techniques, and analyzed overall survival (OS) from first recurrence. Results Among the entire cohort, larger tumor size at first recurrence was associated with poorer survival. The CCNU 01-04 group had similar tumor size as the Bev arms and low Bev crossover (7%). Treatment with Bev was associated with improved survival in patients with large tumor 2D measurements: Median OS for Bev and Bev/CCNU groups were 6.71 mo (n = 27) and 6.97 mo (n = 36) vs 4.03 mo (n = 10) in CCNU 01-04. Analysis by 3D measurement yielded similar results. Interestingly, the CCNU 09-15 group showed the highest survival, likely due to smaller tumor size and crossover to Bev (69%). Conclusion Survival advantage from Bev treatment was observed only among patients with large tumor burden as determined by either 2D or 3D measurement.
Collapse
Affiliation(s)
- Huy Tram Nguyen
- Department of Neurology, University of California, Los Angeles, California, USA
| | - Nhung Nguyen
- Department of Neurology, University of California, Los Angeles, California, USA
| | - Liang Yen Liu
- Department of Neurology, University of California, Los Angeles, California, USA
| | - Laura Dovek
- Department of Neurology, University of California, Los Angeles, California, USA
| | - Daniel Lenchner
- Department of Neurology, University of California, Los Angeles, California, USA
| | - Robert Harris
- Department of Radiological Sciences and Biomedical Physics, University of California, Los Angeles, California, USA
| | - Byram Ozer
- Department of Neurology, University of California, Los Angeles, California, USA
| | | | | | - Myung Shin Sim
- Department of Biomathematics, University of California, Los Angeles, California, USA
| | - Robert Elashoff
- Department of Biomathematics, University of California, Los Angeles, California, USA
| | - Richard Green
- Kaiser Permanente Southern California, Los Angeles, California, USA
| | | | | | - Benjamin Ellingson
- Department of Radiological Sciences and Biomedical Physics, University of California, Los Angeles, California, USA
| | - Albert Lai
- Department of Neurology, University of California, Los Angeles, California, USA
| |
Collapse
|
629
|
Timmons JJ, Preto J, Tuszynski JA, Wong ET. Tubulin's response to external electric fields by molecular dynamics simulations. PLoS One 2018; 13:e0202141. [PMID: 30231050 PMCID: PMC6145594 DOI: 10.1371/journal.pone.0202141] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/27/2018] [Indexed: 02/03/2023] Open
Abstract
Tubulin heterodimers are the building blocks of microtubules and disruption of their dynamics is exploited in the treatment of cancer. Electric fields at certain frequencies and magnitudes are believed to do the same. Here, the tubulin dimer’s response to external electric fields was determined by atomistic simulation. External fields from 50 to 750 kV/cm, applied for 10 ns, caused significant conformational rearrangements that were dependent upon the field’s directionality. Charged and flexible regions, including the α:H1-B2 loop, β:M-loop, and C-termini, were susceptible. Closer inspection of the α:H1-B2 loop in lower strength fields revealed that these effects were consistent and proportional to field strength, and the findings indicate that external electric fields modulate the stability of microtubules through conformational changes to key loops involved in lateral contacts. We also find evidence that tubulin’s curvature and elongation are affected, and external electric fields may bias tubulin towards depolymerization.
Collapse
Affiliation(s)
- Joshua J. Timmons
- Brain Tumor Center & Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jordane Preto
- Department of Physics, University of Alberta, Edmonton, Canada
| | - Jack A. Tuszynski
- Department of Physics, University of Alberta, Edmonton, Canada
- Department of Oncology, University of Alberta, Edmonton, Canada
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi, Torino, Italy
| | - Eric T. Wong
- Brain Tumor Center & Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
630
|
Newton SL, Kalamaha K, Fernandes HD. Temozolomide-induced Aplastic Anemia Treated with Eltrombopag and Granulocyte Colony Stimulating Factor: A Report of a Rare Complication. Cureus 2018; 10:e3329. [PMID: 30473962 PMCID: PMC6248811 DOI: 10.7759/cureus.3329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Temozolomide is an alkylating agent used in the treatment for glioblastoma multiforme (GBM), the most frequent primary malignant brain tumor in adults. Temozolomide was approved in March 2005 for treatment of GBM, with the Stupp protocol (radiotherapy and concomitant use of temozolomide). Despite initial studies demonstrating mild and well-tolerated side effects, several recent reports describe severe hematologic adverse effects associated with temozolomide use. We report the case of a 51-year-old female diagnosed with GBM who received the standard treatment protocol of radiotherapy and concomitant temozolomide. The patient developed prolonged pancytopenia. Bone marrow biopsy demonstrated hypocellular bone marrow with diminished trilineage hematopoiesis, suggestive of drug-induced aplastic anemia. Although temozolomide is regarded as a safe drug with few side effects, severe hematologic toxicities have been reported.
Collapse
Affiliation(s)
- Shauna L Newton
- Internal Medicine, University of North Dakota School of Medicine and Health Sciences, Bismarck, USA
| | - Kadra Kalamaha
- Internal Medicine, University of North Dakota School of Medicine and Health Sciences, Bismarck, USA
| | - Hermina D Fernandes
- Internal Medicine, University of North Dakota School of Medicine and Health Sciences, Bismarck, USA
| |
Collapse
|
631
|
Vecera M, Sana J, Lipina R, Smrcka M, Slaby O. Long Non-Coding RNAs in Gliomas: From Molecular Pathology to Diagnostic Biomarkers and Therapeutic Targets. Int J Mol Sci 2018; 19:ijms19092754. [PMID: 30217088 PMCID: PMC6163683 DOI: 10.3390/ijms19092754] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022] Open
Abstract
Gliomas are the most common malignancies of the central nervous system. Because of tumor localization and the biological behavior of tumor cells, gliomas are characterized by very poor prognosis. Despite significant efforts that have gone into glioma research in recent years, the therapeutic efficacy of available treatment options is still limited, and only a few clinically usable diagnostic biomarkers are available. More and more studies suggest non-coding RNAs to be promising diagnostic biomarkers and therapeutic targets in many cancers, including gliomas. One of the largest groups of these molecules is long non-coding RNAs (lncRNAs). LncRNAs show promising potential because of their unique tissue expression patterns and regulatory functions in cancer cells. Understanding the role of lncRNAs in gliomas may lead to discovery of the novel molecular mechanisms behind glioma biological features. It may also enable development of new solutions to overcome the greatest obstacles in therapy of glioma patients. In this review, we summarize the current knowledge about lncRNAs and their involvement in the molecular pathology of gliomas. A conclusion follows that these RNAs show great potential to serve as powerful diagnostic, prognostic, and predictive biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Marek Vecera
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic.
| | - Jiri Sana
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic.
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Radim Lipina
- Department of Neurosurgery, University Hospital Ostrava, 70852 Ostrava, Czech Republic.
| | - Martin Smrcka
- Department of Neurosurgery, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic.
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.
| |
Collapse
|
632
|
Alphandéry E. Glioblastoma Treatments: An Account of Recent Industrial Developments. Front Pharmacol 2018; 9:879. [PMID: 30271342 PMCID: PMC6147115 DOI: 10.3389/fphar.2018.00879] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/20/2018] [Indexed: 12/28/2022] Open
Abstract
The different drugs and medical devices, which are commercialized or under industrial development for glioblastoma treatment, are reviewed. Their different modes of action are analyzed with a distinction being made between the effects of radiation, the targeting of specific parts of glioma cells, and immunotherapy. Most of them are still at a too early stage of development to firmly conclude about their efficacy. Optune, which triggers antitumor activity by blocking the mitosis of glioma cells under the application of an alternating electric field, seems to be the only recently developed therapy with some efficacy reported on a large number of GBM patients. The need for early GBM diagnosis is emphasized since it could enable the treatment of GBM tumors of small sizes, possibly easier to eradicate than larger tumors. Ways to improve clinical protocols by strengthening preclinical studies using of a broader range of different animal and tumor models are also underlined. Issues related with efficient drug delivery and crossing of blood brain barrier are discussed. Finally societal and economic aspects are described with a presentation of the orphan drug status that can accelerate the development of GBM therapies, patents protecting various GBM treatments, the different actors tackling GBM disease, the cost of GBM treatments, GBM market figures, and a financial analysis of the different companies involved in the development of GBM therapies.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, UMR 7590 CNRS, Sorbonne Universités, UPMC, University Paris 06, Paris, France.,Nanobacterie SARL, Paris, France
| |
Collapse
|
633
|
Diaz P, Horne E, Xu C, Hamel E, Wagenbach M, Petrov RR, Uhlenbruck B, Haas B, Hothi P, Wordeman L, Gussio R, Stella N. Modified carbazoles destabilize microtubules and kill glioblastoma multiform cells. Eur J Med Chem 2018; 159:74-89. [PMID: 30268825 DOI: 10.1016/j.ejmech.2018.09.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/07/2018] [Accepted: 09/09/2018] [Indexed: 11/26/2022]
Abstract
Small molecules that target microtubules (MTs) represent promising therapeutics to treat certain types of cancer, including glioblastoma multiform (GBM). We synthesized modified carbazoles and evaluated their antitumor activity in GBM cells in culture. Modified carbazoles with an ethyl moiety linked to the nitrogen of the carbazole and a carbonyl moiety linked to distinct biaromatic rings exhibited remarkably different killing activities in human GBM cell lines and patient-derived GBM cells, with IC50 values from 67 to >10,000 nM. Measures of the activity of modified carbazoles with tubulin and microtubules coupled to molecular docking studies show that these compounds bind to the colchicine site of tubulin in a unique low interaction space that inhibits tubulin assembly. The modified carbazoles reported here represent novel chemical tools to better understand how small molecules disrupt MT functions and kill devastating cancers such as GBM.
Collapse
Affiliation(s)
- Philippe Diaz
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA; DermaXon LLC, 32 Campus Drive, Missoula, MT, 59812, USA.
| | - Eric Horne
- Stella Therapeutics, Inc., Pacific Northwest Research Institute, 720 Broadway, Seattle, WA, 98122, USA
| | - Cong Xu
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Ernest Hamel
- Screening Technologies Branch (EH) and Computational Drug Development Group (RG), Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Michael Wagenbach
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Ravil R Petrov
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
| | - Benjamin Uhlenbruck
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
| | - Brian Haas
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Parvinder Hothi
- Ivy Center for Advance Brain Tumor Treatment, Swedish Neuroscience Institute, 550 17th Ave, Seattle, WA, 98122, USA
| | - Linda Wordeman
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Rick Gussio
- Screening Technologies Branch (EH) and Computational Drug Development Group (RG), Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Nephi Stella
- Stella Therapeutics, Inc., Pacific Northwest Research Institute, 720 Broadway, Seattle, WA, 98122, USA; Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
634
|
Treatment of Gliomas: A Changing Landscape. Int J Radiat Oncol Biol Phys 2018; 98:255-258. [PMID: 29941232 DOI: 10.1016/j.ijrobp.2017.02.223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 02/27/2017] [Indexed: 11/21/2022]
|
635
|
The Safety of Bevacizumab Administered Shortly after Laser Interstitial Thermal Therapy in Glioblastoma: A Case Series. World Neurosurg 2018; 117:e588-e594. [DOI: 10.1016/j.wneu.2018.06.092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 11/18/2022]
|
636
|
Patel CB, Fazzari E, Chakhoyan A, Yao J, Raymond C, Nguyen H, Manoukian J, Nguyen N, Pope W, Cloughesy TF, Nghiemphu PL, Czernin J, Lai A, Ellingson BM. 18F-FDOPA PET and MRI characteristics correlate with degree of malignancy and predict survival in treatment-naïve gliomas: a cross-sectional study. J Neurooncol 2018; 139:399-409. [PMID: 29679199 PMCID: PMC6092195 DOI: 10.1007/s11060-018-2877-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/16/2018] [Indexed: 10/17/2022]
Abstract
INTRODUCTION To report the potential value of pre-operative 18F-FDOPA PET and anatomic MRI in diagnosis and prognosis of glioma patients. METHODS Forty-five patients with a pathological diagnosis of glioma with pre-operative 18F-FDOPA PET and anatomic MRI were retrospectively examined. The volume of contrast enhancement and T2 hyperintensity on MRI images along with the ratio of maximum 18F-FDOPA SUV in tumor to normal tissue (T/N SUVmax) were measured and used to predict tumor grade, molecular status, and overall survival (OS). RESULTS A significant correlation was observed between WHO grade and: the volume of contrast enhancement (r = 0.67), volume of T2 hyperintensity (r = 0.42), and 18F-FDOPA uptake (r = 0.60) (P < 0.01 for each correlation). The volume of contrast enhancement and 18F-FDOPA T/N SUVmax were significantly higher in glioblastoma (WHO IV) compared with lower grade gliomas (WHO I-III), as well as for high-grade gliomas (WHO III-IV) compared with low-grade gliomas (WHO I-II). Receiver-operator characteristic (ROC) analyses confirmed the volume of contrast enhancement and 18F-FDOPA T/N SUVmax could each differentiate patient groups. No significant differences in 18F-FDOPA uptake were observed by IDH or MGMT status. Multivariable Cox regression suggested age (HR 1.16, P = 0.0001) and continuous measures of 18F-FDOPA PET T/N SUVmax (HR 4.43, P = 0.016) were significant prognostic factors for OS in WHO I-IV gliomas. CONCLUSIONS Current findings suggest a potential role for the use of pre-operative 18F-FDOPA PET in suspected glioma. Increased 18F-FDOPA uptake may not only predict higher glioma grade, but also worse OS.
Collapse
Affiliation(s)
- Chirag B Patel
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Elisa Fazzari
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Huytram Nguyen
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jasmine Manoukian
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Nhung Nguyen
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Whitney Pope
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Phioanh L Nghiemphu
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Johannes Czernin
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Albert Lai
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Benjamin M Ellingson
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.
| |
Collapse
|
637
|
Bamberger SN, Malik CK, Voehler MW, Brown SK, Pan H, Johnson-Salyard TL, Rizzo CJ, Stone MP. Configurational and Conformational Equilibria of N 6-(2-Deoxy-d-erythro-pentofuranosyl)-2,6-diamino-3,4-dihydro-4-oxo-5- N-methylformamidopyrimidine (MeFapy-dG) Lesion in DNA. Chem Res Toxicol 2018; 31:924-935. [PMID: 30169026 DOI: 10.1021/acs.chemrestox.8b00135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The most common lesion in DNA occurring due to clinical treatment with Temozolomide or cellular exposures to other methylating agents is 7-methylguanine (N7-Me-dG). It can undergo a secondary reaction to form N6-(2-deoxy-d-erythro-pentofuranosyl)-2,6-diamino-3,4-dihydro-4-oxo-5- N-methylformamidopyrimidine (MeFapy-dG). MeFapy-dG undergoes epimerization in DNA to produce either α or β deoxyribose anomers. Additionally, conformational rotation around the formyl bond, C5- N5 bond, and glycosidic bond may occur. To characterize and quantitate the mixture of these isomers in DNA, a 13C-MeFapy-dG lesion, in which the CH3 group of the MeFapy-dG was isotopically labeled, was incorporated into the trimer 5'-TXT-3' and the dodecamer 5'-CATXATGACGCT-3' (X = 13C-MeFapy-dG). NMR spectroscopy of both the trimer and dodecamer revealed that the MeFapy-dG lesion exists in single strand DNA as ten configurationally and conformationally discrete species, eight of which may be unequivocally assigned. In the duplex dodecamer, the MeFapy-dG lesion exists as six configurationally and conformationally discrete species. Analyses of NMR data in the single strand trimer confirm that for each deoxyribose anomer, atropisomerism occurs around the C5- N5 bond to produce R a and S a atropisomers. Each atropisomer exhibits geometrical isomerism about the formyl bond yielding E and Z conformations. 1H NMR experiments allow the relative abundances of the species to be determined. For the single strand trimer, the α and β anomers exist in a 3:7 ratio, favoring the β anomer. For the β anomer, with respect to the C5- N5 bond, the R a and S a atropisomers are equally populated. However, the Z geometrical isomer of the formyl moiety is preferred. For the α anomer, the E- S a isomer is present at 12%, whereas all other isomers are present at 5-7%. DNA processing enzymes may differentially recognize different isomers of the MeFapy-dG lesion. Moreover, DNA sequence-specific differences in the populations of configurational and conformational species may modulate biological responses to the MeFapy-dG lesion.
Collapse
Affiliation(s)
- Stephanie N Bamberger
- Department of Chemistry , Vanderbilt University Center for Structural Biology, Vanderbilt Center in Molecular Toxicology, and the Vanderbilt-Ingram Cancer Center, Vanderbilt University , Nashville , Tennessee 37235 , United States
| | - Chanchal K Malik
- Department of Chemistry , Vanderbilt University Center for Structural Biology, Vanderbilt Center in Molecular Toxicology, and the Vanderbilt-Ingram Cancer Center, Vanderbilt University , Nashville , Tennessee 37235 , United States
| | - Markus W Voehler
- Department of Chemistry , Vanderbilt University Center for Structural Biology, Vanderbilt Center in Molecular Toxicology, and the Vanderbilt-Ingram Cancer Center, Vanderbilt University , Nashville , Tennessee 37235 , United States
| | - Summer K Brown
- Department of Chemistry , Vanderbilt University Center for Structural Biology, Vanderbilt Center in Molecular Toxicology, and the Vanderbilt-Ingram Cancer Center, Vanderbilt University , Nashville , Tennessee 37235 , United States
| | - Hope Pan
- Department of Chemistry , Vanderbilt University Center for Structural Biology, Vanderbilt Center in Molecular Toxicology, and the Vanderbilt-Ingram Cancer Center, Vanderbilt University , Nashville , Tennessee 37235 , United States
| | - Tracy L Johnson-Salyard
- Department of Chemistry , Vanderbilt University Center for Structural Biology, Vanderbilt Center in Molecular Toxicology, and the Vanderbilt-Ingram Cancer Center, Vanderbilt University , Nashville , Tennessee 37235 , United States
| | - Carmelo J Rizzo
- Department of Chemistry , Vanderbilt University Center for Structural Biology, Vanderbilt Center in Molecular Toxicology, and the Vanderbilt-Ingram Cancer Center, Vanderbilt University , Nashville , Tennessee 37235 , United States
| | - Michael P Stone
- Department of Chemistry , Vanderbilt University Center for Structural Biology, Vanderbilt Center in Molecular Toxicology, and the Vanderbilt-Ingram Cancer Center, Vanderbilt University , Nashville , Tennessee 37235 , United States
| |
Collapse
|
638
|
Bota DA, Chung J, Dandekar M, Carrillo JA, Kong XT, Fu BD, Hsu FP, Schönthal AH, Hofman FM, Chen TC, Zidovetzki R, Pretto C, Strik A, Schijns VE, Stathopoulos A. Phase II study of ERC1671 plus bevacizumab versus bevacizumab plus placebo in recurrent glioblastoma: interim results and correlations with CD4 + T-lymphocyte counts. CNS Oncol 2018; 7:CNS22. [PMID: 30157683 PMCID: PMC6200061 DOI: 10.2217/cns-2018-0009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aim: ERC1671 is an allogeneic/autologous therapeutic glioblastoma (GBM) vaccine – composed of whole, inactivated tumor cells mixed with tumor cell lysates derived from the patient and three GBM donors. Methods: In this double-blinded, randomized, Phase II study bevacizumab-naive patients with recurrent GBM were randomized to receive either ERC1671 in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) (Leukine® or sargramostim) and cyclophosphamide plus bevacizumab, or placebo plus bevacizumab. Interim results: Median overall survival (OS) of patients treated with ERC1671 plus bevacizumab was 12 months. In the placebo plus bevacizumab group, median OS was 7.5 months. The maximal CD4+ T-lymphocyte count correlated with OS in the ERC1671 but not in the placebo group. Conclusion: The addition of ERC1671/GM-CSF/cyclophosphamide to bevacizumab resulted in a clinically meaningful survival benefit with minimal additional toxicity.
Collapse
Affiliation(s)
- Daniela A Bota
- Department of Neurology, University of California Irvine, Irvine, CA 92868, USA.,Department of Neurological Surgery, University of California Irvine, Irvine, CA 92868, USA.,Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92868, USA
| | - Jinah Chung
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92868, USA
| | - Manisha Dandekar
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92868, USA
| | - Jose A Carrillo
- Department of Neurology, University of California Irvine, Irvine, CA 92868, USA.,Department of Neurological Surgery, University of California Irvine, Irvine, CA 92868, USA.,Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92868, USA
| | - Xiao-Tang Kong
- Department of Neurology, University of California Irvine, Irvine, CA 92868, USA.,Department of Neurological Surgery, University of California Irvine, Irvine, CA 92868, USA.,Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92868, USA
| | - Beverly D Fu
- Department of Neurology, University of California Irvine, Irvine, CA 92868, USA.,Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92868, USA
| | - Frank Pk Hsu
- Department of Neurological Surgery, University of California Irvine, Irvine, CA 92868, USA.,Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92868, USA
| | - Axel H Schönthal
- Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Florence M Hofman
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Thomas C Chen
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Raphael Zidovetzki
- Cell Biology & Neuroscience, University of California, Riverside, CA 92507, USA
| | - Chrystel Pretto
- Epitopoietic Research Corporation, Gembloux, 5032 Isnes, Belgium
| | - Ankie Strik
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Epitopoietic Research Corporation, Gembloux, 5032 Isnes, Belgium
| | - Virgil Ejc Schijns
- Epitopoietic Research Corporation, Gembloux, 5032 Isnes, Belgium.,Cell Biology & Immunology Group, Wageningen University, 6708 Wageningen, The Netherlands
| | - Apostolos Stathopoulos
- Epitopoietic Research Corporation, Gembloux, 5032 Isnes, Belgium.,Cell Biology & Immunology Group, Wageningen University, 6708 Wageningen, The Netherlands.,Department of Neurosurgery, Euroclinics Hospital, 151 21 Athens, Greece
| |
Collapse
|
639
|
Vikram B. Perspectives on potential research benefits from big data efforts in Radiation Oncology. Med Phys 2018; 45:e848-e849. [PMID: 30152535 DOI: 10.1002/mp.13109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Bhadrasain Vikram
- Clinical Radiation Oncology Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NIH), Rockville, MD, USA
| |
Collapse
|
640
|
Dose-dense temozolomide for recurrent high-grade gliomas: a single-center retrospective study. Med Oncol 2018; 35:136. [PMID: 30155806 DOI: 10.1007/s12032-018-1198-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 08/22/2018] [Indexed: 01/10/2023]
Abstract
There are limited treatment modalities after high-grade gliomas recurrence. MGMT depletion modulated by dose-dense temozolomide (ddTMZ) remains a debated therapy for initial TMZ responders. Patients were selected retrospectively from our practice with diagnosis of high-grade gliomas (WHO grade III or IV), and were followed since the start of ddTMZ until death or change of therapy. Twenty-one patients were reviewed, with a median age of 47 (25-61) years and a median of 5.8 (1.5-38.8) cycles of ddTMZ. The majority were males (71.4%). Sixty-six percent received 21 on/28 off ddTMZ schedule, 28.6% daily, and 1 patient received a 7 days on/7 days off schedule. IDH mutation status was available for 18 (85.7%) patients, with 7 (33.3%) IDH mutant and 11 (52.5%) IDH wild type. MGMT methylation was assessed in 6 (28.6%) of the patients, being MGMT methylated in 3 (14.3%) patients, and non-methylated in 3 (14.3%) patients. The majority of patients (57.1%) were receiving ddTMZ in addition to other forms of therapy, including either bevacizumab (38.1%) or tumor-treating fields (TTFields) (19.1%). Overall ddTMZ was well tolerated, with few adverse events reported. The estimated median overall survival after ddTMZ start was 11 months. Median progression-free survival (PFS) was 6 months. Outcomes did not vary between patients receiving ddTMZ alone or those using TTFields or bevacizumab as concomitant therapy, but there was a trend to longer survival with the use of concomitant TTFields. Our results demonstrate benefit of ddTMZ after previous treatment with standard TMZ dosing with no apparent increase in treatment-related toxicities. In summary, ddTMZ should be considered in TMZ responsive patients and warrants further investigation.
Collapse
|
641
|
Therapeutic Immunization against Glioblastoma. Int J Mol Sci 2018; 19:ijms19092540. [PMID: 30150597 PMCID: PMC6163986 DOI: 10.3390/ijms19092540] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/18/2018] [Accepted: 08/23/2018] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma is the most common form of brain cancer in adults that produces severe damage to the brain leading to a very poor survival prognosis. The standard of care for glioblastoma is usually surgery, as well as radiotherapy followed by systemic temozolomide chemotherapy, resulting in a median survival time of about 12 to 15 months. Despite these therapeutic efforts, the tumor returns in the vast majority of patients. When relapsing, statistics suggest an imminent death dependent on the size of the tumor, the Karnofsky Performance Status, and the tumor localization. Following the standard of care, the administration of Bevacizumab, inhibiting the growth of the tumor vasculature, is an approved medicinal treatment option approved in the United States, but not in the European Union, as well as the recently approved alternating electric fields (AEFs) generator NovoTTF/Optune. However, it is clear that regardless of the current treatment regimens, glioma patients continue to have dismal prognosis and novel treatments are urgently needed. Here, we describe different approaches of recently developed therapeutic glioma brain cancer vaccines, which stimulate the patient’s immune system to recognize tumor-associated antigens (TAA) on cancer cells, aiming to instruct the immune system to eventually attack and destroy the brain tumor cells, with minimal bystander damage to normal brain cells. These distinct immunotherapies may target particular glioma TAAs which are molecularly defined, but they may also target broad patient-derived tumor antigen preparations intentionally evoking a very broad polyclonal antitumor immune stimulation.
Collapse
|
642
|
Korshoej AR, Hansen FL, Mikic N, von Oettingen G, Sørensen JCH, Thielscher A. Importance of electrode position for the distribution of tumor treating fields (TTFields) in a human brain. Identification of effective layouts through systematic analysis of array positions for multiple tumor locations. PLoS One 2018; 13:e0201957. [PMID: 30133493 PMCID: PMC6104980 DOI: 10.1371/journal.pone.0201957] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 07/25/2018] [Indexed: 12/30/2022] Open
Abstract
Tumor treating fields (TTFields) is a new modality used for the treatment of glioblastoma. It is based on antineoplastic low-intensity electric fields induced by two pairs of electrode arrays placed on the patient’s scalp. The layout of the arrays greatly impacts the intensity (dose) of TTFields in the pathology. The present study systematically characterizes the impact of array position on the TTFields distribution calculated in a realistic human head model using finite element methods. We investigate systematic rotations of arrays around a central craniocaudal axis of the head and identify optimal layouts for a large range of (nineteen) different frontoparietal tumor positions. In addition, we present comprehensive graphical representations and animations to support the users’ understanding of TTFields. For most tumors, we identified two optimal array positions. These positions varied with the translation of the tumor in the anterior-posterior direction but not in the left-right direction. The two optimal directions were oriented approximately orthogonally and when combining two pairs of orthogonal arrays, equivalent to clinical TTFields therapy, we correspondingly found a single optimum position. In most cases, an oblique layout with the fields oriented at forty-five degrees to the sagittal plane was superior to the commonly used anterior-posterior and left-right combinations of arrays. The oblique configuration may be used as an effective and viable configuration for most frontoparietal tumors. Our results may be applied to assist clinical decision-making in various challenging situations associated with TTFields. This includes situations in which circumstances, such as therapy-induced skin rash, scar tissue or shunt therapy, etc., require layouts alternative to the prescribed. More accurate distributions should, however, be based on patient-specific models. Future work is needed to assess the robustness of the presented results towards variations in conductivity.
Collapse
Affiliation(s)
- Anders Rosendal Korshoej
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade, Aarhus C, Denmark
- Aarhus University, Department of Clinical Medicine, Palle Juul-Jensens Boulevard, Aarhus N, Denmark
- * E-mail:
| | - Frederik Lundgaard Hansen
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade, Aarhus C, Denmark
- Aarhus University, Department of Clinical Medicine, Palle Juul-Jensens Boulevard, Aarhus N, Denmark
| | - Nikola Mikic
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade, Aarhus C, Denmark
| | - Gorm von Oettingen
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade, Aarhus C, Denmark
- Aarhus University, Department of Clinical Medicine, Palle Juul-Jensens Boulevard, Aarhus N, Denmark
| | - Jens Christian Hedemann Sørensen
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade, Aarhus C, Denmark
- Aarhus University, Department of Clinical Medicine, Palle Juul-Jensens Boulevard, Aarhus N, Denmark
| | - Axel Thielscher
- Danish Research Center for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Kettegaards Allé, DK, Hvidovre, Denmark
- Department of Electrical Engineering, Technical University of Denmark, Ørsteds Plads, DK, Kgs. Lyngby, Denmark
| |
Collapse
|
643
|
Wildes TJ, Grippin A, Dyson KA, Wummer BM, Damiani DJ, Abraham RS, Flores CT, Mitchell DA. Cross-talk between T Cells and Hematopoietic Stem Cells during Adoptive Cellular Therapy for Malignant Glioma. Clin Cancer Res 2018; 24:3955-3966. [PMID: 29712687 PMCID: PMC6095818 DOI: 10.1158/1078-0432.ccr-17-3061] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/26/2018] [Accepted: 04/25/2018] [Indexed: 01/05/2023]
Abstract
Purpose: Adoptive T-cell immunotherapy (ACT) has emerged as a viable therapeutic for peripheral and central nervous system (CNS) tumors. In peripheral cancers, optimal efficacy of ACT is reliant on dendritic cells (DCs) in the tumor microenvironment. However, the CNS is largely devoid of resident migratory DCs to function as antigen-presenting cells during immunotherapy. Herein, we demonstrate that cellular interactions between adoptively transferred tumor-reactive T cells and bone marrow-derived hematopoietic stem and progenitor cells (HSPCs) lead to the generation of potent intratumoral DCs within the CNS compartment.Experimental Design: We evaluated HSPC differentiation during ACT in vivo in glioma-bearing hosts and HSPC proliferation and differentiation in vitro using a T-cell coculture system. We utilized FACS, ELISAs, and gene expression profiling to study the phenotype and function of HSPC-derived cells ex vivo and in vivo To demonstrate the impact of HSPC differentiation and function on antitumor efficacy, we performed survival experiments.Results: Transfer of HSPCs with concomitant ACT led to the production of activated CD86+CD11c+MHCII+ cells consistent with DC phenotype and function within the brain tumor microenvironment. These intratumoral DCs largely supplanted abundant host myeloid-derived suppressor cells. We determined that during ACT, HSPC-derived cells in gliomas rely on T-cell-released IFNγ to differentiate into DCs, activate T cells, and reject intracranial tumors.Conclusions: Our data support the use of HSPCs as a novel cellular therapy. Although DC vaccines induce robust immune responses in the periphery, our data demonstrate that HSPC transfer uniquely generates intratumoral DCs that potentiate T-cell responses and promote glioma rejection in situClin Cancer Res; 24(16); 3955-66. ©2018 AACR.
Collapse
Affiliation(s)
- Tyler J Wildes
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Adam Grippin
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Kyle A Dyson
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Brandon M Wummer
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - David J Damiani
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Rebecca S Abraham
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Catherine T Flores
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida.
| | - Duane A Mitchell
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida.
| |
Collapse
|
644
|
Koh HK, Seo SY, Kim JH, Kim HJ, Chie EK, Kim SK, Kim IH. Disulfiram, a Re-positioned Aldehyde Dehydrogenase Inhibitor, Enhances Radiosensitivity of Human Glioblastoma Cells In Vitro. Cancer Res Treat 2018; 51:696-705. [PMID: 30121967 PMCID: PMC6473295 DOI: 10.4143/crt.2018.249] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/10/2018] [Indexed: 11/21/2022] Open
Abstract
Purpose Glioblastoma, the most common brain tumor in adults, has poor prognosis. The purpose of this study was to determine the effect of disulfiram (DSF), an aldehyde dehydrogenase inhibitor, on in vitro radiosensitivity of glioblastoma cells with different methylation status of O6-methylguanine-DNA methyltransferase (MGMT) promoter and the underlying mechanism of such effect. Materials and Methods Five human glioblastoma cells (U138MG, T98G, U251MG, U87MG, and U373MG) and one normal human astrocyte (NHA) cell were cultured and treated with DSF or 6MV X-rays (0, 2, 4, 6, and 8 Gy). For combined treatment, cells were treated with DSF before irradiation. Surviving fractions fit from cell survival based on colony forming ability. Apoptosis, DNA damage repair, and cell cycle distributionwere assayed bywestern blot for cleaved caspase-3, γH2AX staining, and flow cytometry, respectively. Results DSF induced radiosensitization in most of the glioblastoma cells, especially, in the cells with radioresistance as wildtype unmethylated promoter (MGMT-wt), but did not in normal NHA cell. DSF augmented or induced cleavage of caspase-3 in all cells after irradiation. DSF inhibited repair of radiation-induced DNA damage in MGMT-wt cells, but not in cells with methylated MGMT promoter. DSF abrogated radiation-induced G2/M arrest in T98G and U251MG cells. Conclusion Radiosensitivity of glioblastoma cells were preferentially enhanced by pre-irradiation DSF treatment compared to normal cell, especially radioresistant cells such as MGMT-wt cells. Induction of apoptosis or inhibition of DNA damage repair may underlie DSF-induced radiosensitization. Clinical benefit of combining DSF with radiotherapy should be investigated in the future.
Collapse
Affiliation(s)
- Hyeon Kang Koh
- Department of Radiation Oncology, Konkuk University Medical Center, Seoul, Korea.,Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Yeon Seo
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Jin Ho Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Eui Kyu Chie
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Korea
| | - Il Han Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
645
|
Glioblastoma radiomics: can genomic and molecular characteristics correlate with imaging response patterns? Neuroradiology 2018; 60:1043-1051. [PMID: 30094640 DOI: 10.1007/s00234-018-2060-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE For glioblastoma (GBM), imaging response (IR) or pseudoprogression (PSP) is frequently observed after chemoradiation and may connote a favorable prognosis. With tumors categorized by the Cancer Genome Atlas Project (mesenchymal, classical, neural, and proneural) and by methylguanine-methyltransferase (MGMT) methylation status, we attempted to determine if certain genomic or molecular subtypes of GBM were specifically associated with IR or PSP. METHODS Patients with GBM treated at two institutions were reviewed. Kaplan-Meier method was used to estimate overall survival (OS) and progression-free survival (PFS). Mantel-cox test determined effect of IR and PSP on OS and PFS. Fisher's exact test was utilized to correlate IR and PSP with genomic subtypes and MGMT status. RESULTS Eighty-two patients with GBM were reviewed. The median OS and PFS were 17.9 months and 8.9 months. IR was observed in 28 (40%) and was associated with improved OS (median 29.4 vs 14.5 months p < 0.01) and PFS (median 17.7 vs 5.5 months, p < 0.01). PSP was observed in 14 (19.2%) and trended towards improved PFS (15.0 vs 7.7 months p = 0.08). Tumors with a proneural component had a higher rate of IR compared to those without a proneural component (IR 60% vs 28%; p = 0.03). MGMT methylation was associated with IR (58% vs 24%, p = 0.032), but not PSP (34%, p = 0.10). CONCLUSION IR is associated with improved OS and PFS. The proneural subtype and MGMT methylated tumors had higher rates of IR.
Collapse
|
646
|
Abstract
PURPOSE OF REVIEW This article reviews the standard treatment for high-grade gliomas, with a focus on promising new strategies and response assessment. RECENT FINDINGS The new World Health Organization (WHO) classification of central nervous system tumors classifies high-grade gliomas based on molecular markers that are of prognostic and therapeutic significance. The addition of chemotherapy, specifically procarbazine, CCNU (lomustine), and vincristine, to radiation in newly diagnosed 1p/19q codeleted anaplastic oligodendrogliomas doubled overall survival. The US Food and Drug Administration (FDA) recently approved the addition of tumor treating fields to adjuvant temozolomide after radiation with concurrent temozolomide in newly diagnosed glioblastoma. A phase3 trial for recurrent glioblastoma did not show an overall survival benefit for the addition of bevacizumab to lomustine compared to lomustine alone. Current efforts are focused on the development of novel treatment approaches, including molecular targeted agents and immunotherapies. SUMMARY Surgery, radiation, and chemotherapy remain the standard treatment options for patients with high-grade gliomas. Despite aggressive treatment, these tumors progress, and overall outcomes have not changed much in the past decade. However, our understanding of the disease is improving, and newer therapies appear promising.
Collapse
|
647
|
Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet 2018; 392:432-446. [PMID: 30060998 DOI: 10.1016/s0140-6736(18)30990-5] [Citation(s) in RCA: 867] [Impact Index Per Article: 123.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/05/2018] [Accepted: 04/23/2018] [Indexed: 12/11/2022]
Abstract
Primary CNS tumours refer to a heterogeneous group of tumours arising from cells within the CNS, and can be benign or malignant. Malignant primary brain tumours remain among the most difficult cancers to treat, with a 5 year overall survival no greater than 35%. The most common malignant primary brain tumours in adults are gliomas. Recent advances in molecular biology have improved understanding of glioma pathogenesis, and several clinically significant genetic alterations have been described. A number of these (IDH, 1p/19q codeletion, H3 Lys27Met, and RELA-fusion) are now combined with histology in the revised 2016 WHO classification of CNS tumours. It is likely that understanding such molecular alterations will contribute to the diagnosis, grading, and treatment of brain tumours. This progress in genomics, along with significant advances in cancer and CNS immunology, has defined a new era in neuro-oncology and holds promise for diagntic and therapeutic improvement. The challenge at present is to translate these advances into effective treatments. Current efforts are focused on developing molecular targeted therapies, immunotherapies, gene therapies, and novel drug-delivery technologies. Results with single-agent therapies have been disappointing so far, and combination therapies seem to be required to achieve a broad and durable antitumour response. Biomarker-targeted clinical trials could improve efficiencies of therapeutic development.
Collapse
Affiliation(s)
- Sarah Lapointe
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Arie Perry
- Division of Neuropathology, Department of Pathology, University of California, San Francisco, CA, USA
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, CA, USA.
| |
Collapse
|
648
|
Gouazé-Andersson V, Ghérardi MJ, Lemarié A, Gilhodes J, Lubrano V, Arnauduc F, Cohen-Jonathan Moyal E, Toulas C. FGFR1/FOXM1 pathway: a key regulator of glioblastoma stem cells radioresistance and a prognosis biomarker. Oncotarget 2018; 9:31637-31649. [PMID: 30167084 PMCID: PMC6114977 DOI: 10.18632/oncotarget.25827] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma are known to be aggressive and therapy-resistant tumors, due to the presence of glioblastoma stem cells inside this heterogeneous tumor. We investigate here the involvement of FGFR1 in glioblastoma stem-like cells (GSLC) radioresistance mechanisms. We first demonstrated that the survival after irradiation was significantly diminished in FGFR1-silenced (FGFR1-) GSLC compared to control GSLC. The transcriptome analysis of GSLCs FGFR1(-) showed that FOX family members are differentially regulated by FGFR1 inhibition, particularly with an upregulation of FOXN3 and a downregulation of FOXM1. GSLC survival after irradiation was significantly increased after FOXN3 silencing and decreased after FOXM1 inhibition, showing opposite effects of FGFR1/FOX family members on cell response to ionizing radiation. Silencing FGFR1 or FOXM1 downregulated genes involved in mesenchymal transition such as GLI2, TWIST1, and ZEB1 in glioblastoma stem-like cells. It also dramatically reduced GSLC migration. Databases analysis confirmed that the combined expression of FGFR1/FOXM1/MELK/GLI2/ZEB1/TWIST1 is significantly associated with patients overall survival after chemo-radiotherapy treatment. All these results, associated with our previous conduced ones with differentiated cells, clearly established that FGFR1-FOXM1 dependent glioblastoma stem-like cells radioresistance pathway is a central actor of GBM treatment resistance and a key target to inhibit in the aim to increase the sensitivity of GBM to the radiotherapy.
Collapse
Affiliation(s)
- Valérie Gouazé-Andersson
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Marie-Julie Ghérardi
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Anthony Lemarié
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Julia Gilhodes
- Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| | - Vincent Lubrano
- CHU PURPAN-Pavillon Baudot, Place du Dr Baylac, Toulouse-Cedex 3, 31024, France
| | - Florent Arnauduc
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Elizabeth Cohen-Jonathan Moyal
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France.,Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| | - Christine Toulas
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France.,Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| |
Collapse
|
649
|
Angiogenesis inhibition on glioblastoma multiforme cell lines (U-87 MG and T98G) by AT-101. JOURNAL OF ONCOLOGICAL SCIENCES 2018. [DOI: 10.1016/j.jons.2018.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
650
|
Abstract
Brain tumors are common, requiring general medical providers to have a basic understanding of their diagnosis and management. The most prevalent brain tumors are intracranial metastases from systemic cancers, meningiomas, and gliomas, specifically, glioblastoma. Central nervous system metastases may occur anywhere along the neuroaxis, and require complex multidisciplinary care with neurosurgery, radiation oncology, and medical oncology. Meningiomas are tumors of the meninges, mostly benign and often managed by surgical resection, with radiation therapy and chemotherapy reserved for high-risk or refractory disease. Glioblastoma is the most common and aggressive malignant primary brain tumor, with a limited response to standard-of-care concurrent chemoradiation. The new classification of gliomas relies on molecular features, as well as histology, to arrive at an "integrated diagnosis" that better captures prognosis. This manuscript will review the most common brain tumors with an emphasis on their diagnosis, oncologic management, and management of medical complications.
Collapse
Affiliation(s)
- J Ricardo McFaline-Figueroa
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Mass; Department of Neurology, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Department of Neurology, Massachusetts General Hospital, Boston
| | - Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Mass; Department of Neurology, Brigham and Women's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|