1
|
Wang JZ, Landry AP, Raleigh DR, Sahm F, Walsh KM, Goldbrunner R, Yefet LS, Tonn JC, Gui C, Ostrom QT, Barnholtz-Sloan J, Perry A, Ellenbogen Y, Hanemann CO, Jungwirth G, Jenkinson MD, Tabatabai G, Mathiesen TI, McDermott MW, Tatagiba M, la Fougère C, Maas SLN, Galldiks N, Albert NL, Brastianos PK, Ehret F, Minniti G, Lamszus K, Ricklefs FL, Schittenhelm J, Drummond KJ, Dunn IF, Pathmanaban ON, Cohen-Gadol AA, Sulman EP, Tabouret E, Le Rhun E, Mawrin C, Moliterno J, Weller M, Bi W(L, Gao A, Yip S, Niyazi M, Aldape K, Wen PY, Short S, Preusser M, Nassiri F, Zadeh G. Meningioma: International Consortium on Meningiomas consensus review on scientific advances and treatment paradigms for clinicians, researchers, and patients. Neuro Oncol 2024; 26:1742-1780. [PMID: 38695575 PMCID: PMC11449035 DOI: 10.1093/neuonc/noae082] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
Meningiomas are the most common primary intracranial tumors in adults and are increasing in incidence due to the aging population and increased access to neuroimaging. While most exhibit nonmalignant behavior, a subset of meningiomas are biologically aggressive and are associated with treatment resistance, resulting in significant neurologic morbidity and even mortality. In recent years, meaningful advances in our understanding of the biology of these tumors have led to the incorporation of molecular biomarkers into their grading and prognostication. However, unlike other central nervous system (CNS) tumors, a unified molecular taxonomy for meningiomas has not yet been established and remains an overarching goal of the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy-Not Official World Health Organization (cIMPACT-NOW) working group. Additionally, clinical equipoise still remains on how specific meningioma cases and patient populations should be optimally managed. To address these existing gaps, members of the International Consortium on Meningiomas including field-leading experts, have prepared this comprehensive consensus narrative review directed toward clinicians, researchers, and patients. Included in this manuscript are detailed overviews of proposed molecular classifications, novel biomarkers, contemporary treatment strategies, trials on systemic therapies, health-related quality-of-life studies, and management strategies for unique meningioma patient populations. In each section, we discuss the current state of knowledge as well as ongoing clinical and research challenges to road map future directions for further investigation.
Collapse
Affiliation(s)
- Justin Z Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Alexander P Landry
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - David R Raleigh
- Department of Radiation Oncology, Neurological Surgery, and Pathology, University of California San Francisco, San Francisco, California, USA
| | - Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kyle M Walsh
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Roland Goldbrunner
- Center of Neurosurgery, Department of General Neurosurgery, University of Cologne, Cologne, Germany
| | - Leeor S Yefet
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Jörg C Tonn
- Department of Neurosurgery, University Hospital Munich LMU, Munich, Germany
| | - Chloe Gui
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Quinn T Ostrom
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Jill Barnholtz-Sloan
- Center for Biomedical Informatics & Information Technology (CBIIT), National Cancer Institute, Bethesda, Maryland, USA
- Trans Divisional Research Program (TDRP), Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute, Bethesda, Maryland, USA
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
| | - Arie Perry
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Yosef Ellenbogen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - C Oliver Hanemann
- Peninsula Schools of Medicine, University of Plymouth University, Plymouth, UK
| | - Gerhard Jungwirth
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University, Heidelberg, Germany
| | - Michael D Jenkinson
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
- Institute of Translational Medicine, University of Liverpool, UK
| | - Ghazaleh Tabatabai
- Department of Neurology and Interdisciplinary Neuro-Oncology, University Hospital Tübingen, Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies,” Eberhard Karls University Tübingen, Tübingen, Germany
- Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Tübingen, Germany
| | - Tiit I Mathiesen
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael W McDermott
- Division of Neuroscience, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
- Miami Neuroscience Institute, Baptist Health of South Florida, Miami, Florida, USA
| | - Marcos Tatagiba
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
- Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tübingen, Germany
- Cluster of Excellence (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies,” Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sybren L N Maas
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (IMN-3), Research Center Juelich, Juelich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany
| | - Priscilla K Brastianos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Felix Ehret
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Giuseppe Minniti
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | - Katrin Lamszus
- Laboratory for Brain Tumor Biology, University Hospital Eppendorf, Hamburg, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Schittenhelm
- Department of Neuropathology, University Hospital Tübingen, Eberhard-Karls-University Tübingen, Tübingen, Germany
- Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Tübingen, Germany
| | - Katharine J Drummond
- Department of Neurosurgery, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Ian F Dunn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Omar N Pathmanaban
- Division of Neuroscience and Experimental Psychology, Manchester Centre for Clinical Neurosciences, Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester, UK
| | - Aaron A Cohen-Gadol
- Department of Neurological Surgery, Indiana University, Indianapolis, Indiana, USA
| | - Erik P Sulman
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Emeline Tabouret
- CNRS, INP, Inst Neurophysiopathol, Aix-Marseille University, Marseille, France
| | - Emelie Le Rhun
- Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Christian Mawrin
- Department of Neuropathology, University Hospital Magdeburg, Magdeburg, Germany
| | - Jennifer Moliterno
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Weller
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Wenya (Linda) Bi
- Department of Neurosurgery, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew Gao
- Department of Laboratory Medicine and Pathobiology, University Health Network, Toronto, Ontario, Canada
| | - Stephen Yip
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Radiation Oncology, University Hospital, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany
| | - Maximilian Niyazi
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, University Hospital Tübingen, Tübingen, Germany
| | | | - Kenneth Aldape
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Patrick Y Wen
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Short
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds, UK
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Farshad Nassiri
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Tomanelli M, Florio T, Vargas GC, Pagano A, Modesto P. Domestic Animal Models of Central Nervous System Tumors: Focus on Meningiomas. Life (Basel) 2023; 13:2284. [PMID: 38137885 PMCID: PMC10744527 DOI: 10.3390/life13122284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/09/2023] [Indexed: 12/24/2023] Open
Abstract
Intracranial primary tumors (IPTs) are aggressive forms of malignancies that cause high mortality in both humans and domestic animals. Meningiomas are frequent adult IPTs in humans, dogs, and cats, and both benign and malignant forms cause a decrease in life quality and survival. Surgery is the primary therapeutic approach to treat meningiomas, but, in many cases, it is not resolutive. The chemotherapy and targeted therapy used to treat meningiomas also display low efficacy and many side effects. Therefore, it is essential to find novel pharmacological approaches to increase the spectrum of therapeutic options for meningiomas. This review analyzes the similarities between human and domestic animal (dogs and cats) meningiomas by evaluating the molecular and histological characteristics, diagnosis criteria, and treatment options and highlighting possible research areas to identify novel targets and pharmacological approaches, which are useful for the diagnosis and therapy of this neoplasia to be used in human and veterinary medicine.
Collapse
Affiliation(s)
- Michele Tomanelli
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy; (G.C.V.); (A.P.)
| | - Tullio Florio
- Pharmacology Section, Department of Internal Medicine (DIMI), University of Genova, 16126 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Gabriela Coronel Vargas
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy; (G.C.V.); (A.P.)
| | - Aldo Pagano
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy; (G.C.V.); (A.P.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Paola Modesto
- National Reference Center for Veterinary and Comparative Oncology, Veterinary Medical Research Institute for Piemonte, Liguria and Valle d’Aosta, 10154 Torino, Italy
| |
Collapse
|
3
|
Azab MA, Cole K, Earl E, Cutler C, Mendez J, Karsy M. Medical Management of Meningiomas. Neurosurg Clin N Am 2023; 34:319-333. [PMID: 37210123 DOI: 10.1016/j.nec.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Meningiomas represent the most common type of benign tumor of the extra-axial compartment. Although most meningiomas are benign World Health Organization (WHO) grade 1 lesions, the increasingly prevalent of WHO grade 2 lesion and occasional grade 3 lesions show worsened recurrence rates and morbidity. Multiple medical treatments have been evaluated but show limited efficacy. We review the status of medical management in meningiomas, highlighting successes and failures of various treatment options. We also explore newer studies evaluating the use of immunotherapy in management.
Collapse
Affiliation(s)
- Mohammed A Azab
- Biomolecular Sciences Graduate Program, Boise State University, 1910 University Drive, Boise, ID 83725, USA
| | - Kyril Cole
- School of Medicine, University of Utah, 30 North 1900 East, Salt Lake City, UT 84132, USA
| | - Emma Earl
- School of Medicine, University of Utah, 30 North 1900 East, Salt Lake City, UT 84132, USA
| | - Chris Cutler
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 N Green Bay Rd., North Chicago, IL 60064, USA
| | - Joe Mendez
- Department of Neurosurgery, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Dr., Salt Lake City, UT 84112, USA
| | - Michael Karsy
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, 175 North Medical Drive East, Salt Lake City, UT 84132, USA.
| |
Collapse
|
4
|
Li Y, Drappatz J. Advances in the systemic therapy for recurrent meningiomas and the challenges ahead. Expert Rev Neurother 2023; 23:995-1004. [PMID: 37695700 DOI: 10.1080/14737175.2023.2254498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023]
Abstract
INTRODUCTION Meningiomas represent the most common primary neoplasms of the central nervous system (CNS). 20% present with atypical (WHO grade II) or malignant (grade III) meningiomas, which show aggressive biologic behavior and high recurrence. Although surgical resection and radiation therapy are the primary treatment options for these tumors, there is a subgroup of patients who do not respond well to or are poor candidates for these approaches, leading to the exploration of systemic therapies as an alternative. AREAS COVERED The literature on different therapeutic groups of systemic drugs for recurrent meningiomas is reviewed, with a focus on the different molecular targets. Past and current ongoing clinical trials are also discussed. EXPERT OPINION To date, there is no recognized treatment that has demonstrated a substantial increase in progression-free or overall survival rates. Nonetheless, therapies targeting anti-VEGF have exhibited more encouraging results in general. The examination of genomic and epigenomic traits of meningiomas, along with the integration of molecular markers into the latest WHO tumor grading system, has provided valuable insights. This has opened avenues for exploring numerous intracellular and extracellular pathways, as well as mutations, that have been targeted in ongoing clinical trials.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology and Medicine, Division of Hematology and Oncology, Center for Neuro-Oncology, Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jan Drappatz
- Department of Neurology and Medicine, Division of Hematology and Oncology, Center for Neuro-Oncology, Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Ijad N, Dahal A, Kim AE, Wakimoto H, Juratli TA, Brastianos PK. Novel Systemic Approaches for the Management of Meningiomas. Neurosurg Clin N Am 2023; 34:447-454. [DOI: 10.1016/j.nec.2023.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
|
6
|
Graillon T, Tabouret E, Salgues B, Horowitz T, Padovani L, Appay R, Farah K, Dufour H, Régis J, Guedj E, Barlier A, Chinot O. Innovative treatments for meningiomas. Rev Neurol (Paris) 2023; 179:449-463. [PMID: 36959063 DOI: 10.1016/j.neurol.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/25/2023]
Abstract
Multi-recurrent high-grade meningiomas remain an unmet medical need in neuro-oncology when iterative surgeries and radiation therapy sessions fail to control tumor growth. Nevertheless, the last 10years have been marked by multiple advances in the comprehension of meningioma tumorigenesis via the discovery of new driver mutations, the identification of activated intracellular signaling pathways, and DNA methylation analyses, providing multiple potential therapeutic targets. Today, Anti-VEGF and mTOR inhibitors are the most used and probably the most active drugs in aggressive meningiomas. Peptide radioactive radiation therapy aims to target SSTR2A receptors, which are strongly expressed in meningiomas, but have an insufficient effect in most aggressive meningiomas, requiring the development of new techniques to increase the dose applied to the tumor. Based on the multiple potential intracellular targets, multiple targeted therapy clinical trials targeting Pi3K-Akt-mTOR and MAP kinase pathways as well as cell cycle and particularly, cyclin D4-6 are ongoing. Recently discovered driver mutations, SMO, Akt, and PI3KCA, offer new targets but are mostly observed in benign meningiomas, limiting their clinical relevance mainly to rare aggressive skull base meningiomas. Therefore, NF2 mutation remains the most frequent mutation and main challenging target in high-grade meningioma. Recently, inhibitors of focal adhesion kinase (FAK), which is involved in tumor cell adhesion, were tested in a phase 2 clinical trial with interesting but insufficient activity. The Hippo pathway was demonstrated to interact with NF2/Merlin and could be a promising target in NF2-mutated meningiomas with ongoing multiple preclinical studies and a phase 1 clinical trial. Recent advances in immune landscape comprehension led to the proposal of the use of immunotherapy in meningiomas. Except in rare cases of MSH2/6 mutation or high tumor mass burden, the activity of PD-1 inhibitors remains limited; however, its combination with various radiation therapy modalities is particularly promising. On the whole, therapeutic management of high-grade meningiomas is still challenging even with multiple promising therapeutic targets and innovations.
Collapse
Affiliation(s)
- T Graillon
- Aix-Marseille University, AP-HM, Inserm, MMG, Neurosurgery department, La Timone Hospital, Marseille, France.
| | - E Tabouret
- Aix-Marseille University, AP-HM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service de Neurooncologie, Marseille, France
| | - B Salgues
- Nuclear Medicine Department, Groupe Hospitalier Pitié-Salpêtrière-Charles-Foix, Assistance publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - T Horowitz
- AP-HM, CNRS, centrale Marseille, Institut Fresnel, Timone Hospital, CERIMED, Nuclear Medicine Department, Aix-Marseille University, Marseille, France
| | - L Padovani
- AP-HM, Timone Hospital, Radiotherapy Department, Marseille, France
| | - R Appay
- AP-HM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France; Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - K Farah
- Aix-Marseille University, Institut de Neurosciences des Systèmes, UMR Inserm 1106, Functional Neurosurgery and Radiosurgery, Timone University Hospital, Marseille, France
| | - H Dufour
- Aix-Marseille University, AP-HM, Inserm, MMG, Neurosurgery department, La Timone Hospital, Marseille, France
| | - J Régis
- Aix-Marseille University, Institut de Neurosciences des Systèmes, UMR Inserm 1106, Functional Neurosurgery and Radiosurgery, Timone University Hospital, Marseille, France
| | - E Guedj
- AP-HM, CNRS, centrale Marseille, Institut Fresnel, Timone Hospital, CERIMED, Nuclear Medicine Department, Aix-Marseille University, Marseille, France
| | - A Barlier
- Aix-Marseille University, AP-HM, Inserm, MMG, Laboratory of Molecular Biology Hospital La Conception, Marseille, France
| | - O Chinot
- Aix-Marseille University, AP-HM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service de Neurooncologie, Marseille, France
| |
Collapse
|
7
|
Youngblood MW, Tran AN, Wang W, An S, Scholtens D, Zhang L, O’Shea K, Pokorny JL, Magill ST, Sachdev S, Lukas RV, Ahmed A, Unruh D, Walshon J, McCortney K, Wang Y, Baran A, Sahm F, Aldape K, Chandler JP, David James C, Heimberger AB, Horbinski C. Docetaxel targets aggressive methylation profiles and serves as a radiosensitizer in high-risk meningiomas. Neuro Oncol 2023; 25:508-519. [PMID: 35976058 PMCID: PMC10013641 DOI: 10.1093/neuonc/noac206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Meningioma is the most common primary intracranial tumor in adults. A subset of these tumors recur and invade the brain, even after surgery and radiation, resulting in significant disability. There is currently no standard-of-care chemotherapy for meningiomas. As genomic DNA methylation profiling can prognostically stratify these lesions, we sought to determine whether any existing chemotherapies might be effective against meningiomas with high-risk methylation profiles. METHODS A previously published dataset of meningioma methylation profiles was used to screen for clinically significant CpG methylation events and associated cellular pathways. Based on these results, patient-derived meningioma cell lines were used to test candidate drugs in vitro and in vivo, including efficacy in conjunction with radiotherapy. RESULTS We identified 981 genes for which methylation of mapped CpG sites was related to progression-free survival in meningiomas. Associated molecular pathways were cross-referenced with FDA-approved cancer drugs, which nominated Docetaxel as a promising candidate for further preclinical analyses. Docetaxel arrested growth in 17 meningioma cell sources, representing all tumor grades, with a clinically favorable IC50 values ranging from 0.3 nM to 10.7 mM. The inhibitory effects of this medication scaled with tumor doubling time, with maximal benefit in fast-growing lesions. The combination of Docetaxel and radiation therapy increased markers of apoptosis and double-stranded DNA breaks, and extended the survival of mice engrafted with meningioma cells relative to either modality alone. CONCLUSIONS Global patterns of DNA methylation may be informative for the selection of chemotherapies against meningiomas, and existing drugs may enhance radiation sensitivity in high-risk cases.
Collapse
Affiliation(s)
- Mark W Youngblood
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Anh N Tran
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Wenxia Wang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Shejuan An
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Denise Scholtens
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Lyndsee Zhang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kaitlyn O’Shea
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jenny L Pokorny
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Stephen T Magill
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sean Sachdev
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Rimas V Lukas
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Atique Ahmed
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dusten Unruh
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jordain Walshon
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yufen Wang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Aneta Baran
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Felix Sahm
- Department of Neuropathology, University of Heidelberg and DKFZ, Heidelberg, Germany
| | - Kenneth Aldape
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - James P Chandler
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Amy B Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Craig Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
8
|
Exploring the Past, Present, and Future of Anti-Angiogenic Therapy in Glioblastoma. Cancers (Basel) 2023; 15:cancers15030830. [PMID: 36765787 PMCID: PMC9913517 DOI: 10.3390/cancers15030830] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Glioblastoma, a WHO grade IV astrocytoma, constitutes approximately half of malignant tumors of the central nervous system. Despite technological advancements and aggressive multimodal treatment, prognosis remains dismal. The highly vascularized nature of glioblastoma enables the tumor cells to grow and invade the surrounding tissue, and vascular endothelial growth factor-A (VEGF-A) is a critical mediator of this process. Therefore, over the past decade, angiogenesis, and more specifically, the VEGF signaling pathway, has emerged as a therapeutic target for glioblastoma therapy. This led to the FDA approval of bevacizumab, a monoclonal antibody designed against VEGF-A, for treatment of recurrent glioblastoma. Despite the promising preclinical data and its theoretical effectiveness, bevacizumab has failed to improve patients' overall survival. Furthermore, several other anti-angiogenic agents that target the VEGF signaling pathway have also not demonstrated survival improvement. This suggests the presence of other compensatory angiogenic signaling pathways that surpass the anti-angiogenic effects of these agents and facilitate vascularization despite ongoing VEGF signaling inhibition. Herein, we review the current state of anti-angiogenic agents, discuss potential mechanisms of anti-angiogenic resistance, and suggest potential avenues to increase the efficacy of this therapeutic approach.
Collapse
|
9
|
Shahbandi A, Shah DS, Hadley CC, Patel AJ. The Role of Pharmacotherapy in Treatment of Meningioma: A Systematic Review. Cancers (Basel) 2023; 15:483. [PMID: 36672431 PMCID: PMC9856307 DOI: 10.3390/cancers15020483] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/14/2023] Open
Abstract
The safety and efficacy of various pharmacotherapeutic regimens on refractory meningiomas have been the focus of investigations. We present a comprehensive review of the previous efforts and the current state of ongoing clinical trials. A PRISMA-compliant review of the MEDLINE and ClinicalTrial.gov databases of the National Library of Medicine were performed. The primary outcomes of interest for included articles were radiographic response, overall survival, progression-free survival, six-month progression-free survival, and adverse events. Overall, 34 completed trials and 27 ongoing clinical trials were eligible. Six-month progression-free survival was reported in 6-100% of patients in the completed studies. Hematological disorders were the most common adverse events. Of the ongoing clinical trials identified, nine studies are phase I clinical trials, eleven are phase II trials, two are phase I and II trials, one is phase II and III, and two trials do not have a designated phase. Currently, there is no effective chemotherapy for refractory or recurrent meningiomas. Several promising targeted agents have been developed and are currently being investigated in the hope of identifying novel therapeutic strategies for the treatment of this pathology.
Collapse
Affiliation(s)
- Ataollah Shahbandi
- Department of Neurological Surgery, School of Medicine, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Darsh S. Shah
- Department of Neurological Surgery, Dell Medical School, Austin, TX 78712, USA
| | - Caroline C. Hadley
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Akash J. Patel
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
| |
Collapse
|
10
|
Mair MJ, Berghoff AS, Brastianos PK, Preusser M. Emerging systemic treatment options in meningioma. J Neurooncol 2023; 161:245-258. [PMID: 36181606 PMCID: PMC9989003 DOI: 10.1007/s11060-022-04148-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/25/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Meningiomas are the most frequently diagnosed intracranial neoplasms. Usually, they are treated by surgical resection in curative intent. Radiotherapy and stereotactic radiosurgery are commonly applied in the adjuvant setting in newly diagnosed atypical (CNS WHO grade 2), and anaplastic (CNS WHO grade 3) meningioma, especially if gross total resection is not feasible, and in recurrent cases. Conversely, the evidence for pharmacotherapy in meningioma is scarce. METHODS The available literature of systemic treatment in meningioma was screened using PubMed, and ongoing clinical trials were explored using ClinicalTrials.gov. RESULTS Classical cytotoxic agents, somatostatin analogs, and antihormone treatments have shown only limited efficacy. In contrast, tyrosine kinase inhibitors and monoclonal antibodies, especially those targeting angiogenic signaling such as sunitinib and bevacizumab, have shown promising antitumoral activity in small phase 2 trials. Moreover, results of recent landmark studies on (epi-)genetic alterations in meningioma revealed potential therapeutic targets which are currently under investigation. These include inhibitors of mammalian target of rapamycin (mTOR), focal adhesion kinase (FAK), cyclin-dependent kinases (CDK), phosphoinositide-3-kinase (PI3K), sonic hedgehog signaling, and histone deacetylases. In addition, clinical trials evaluating immune checkpoint inhibitors such as ipilimumab, nivolumab, pembrolizumab and avelumab are currently being conducted and early results suggest clinically meaningful responses in a subset of patients. CONCLUSIONS There is a paucity of high-level evidence on systemic treatment options in meningioma. However, interesting novel treatment targets have been identified in the last decade. Positive signals of anti-angiogenic agents, genomically targeted agents and immunotherapy in early phase trials should be confirmed in large prospective controlled trials.
Collapse
Affiliation(s)
- Maximilian J Mair
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria
| | - Priscilla K Brastianos
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
11
|
Kawamura Y, Hua L, Gurtner A, Wong E, Kiyokawa J, Shah N, Gorham J, Wakimoto H, Rabkin SD, Martuza RL, Wakimoto H. Histone deacetylase inhibitors enhance oncolytic herpes simplex virus therapy for malignant meningioma. Biomed Pharmacother 2022; 155:113843. [PMID: 36271587 PMCID: PMC9590235 DOI: 10.1016/j.biopha.2022.113843] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022] Open
Abstract
Approximately 20% of meningiomas are not benign (higher grade) and tend to relapse after surgery and radiation therapy. Malignant (anaplastic) meningioma (MM) is a minor subset of high-grade meningioma that is lethal with no effective treatment options currently. Oncolytic herpes simplex virus (oHSV) is a powerful anti-cancer modality that induces both direct cell death and anti-tumor immunity, and has shown activity in preclinical models of MM. However, clinically meaningful efficacy will likely entail rational mechanistic combination approaches. We here show that epigenome modulator histone deacetylase inhibitors (HDACi) increase anti-cancer effects of oHSV in human MM models, IOMM-Lee (NF2 wild-type) and CH157 (NF2 mutant). Minimally toxic, sub-micromolar concentrations of pan-HDACi, Trichostatin A and Panobinostat, substantively increased the infectability and spread of oHSV G47Δ within MM cells in vitro, resulting in enhanced oHSV-mediated killing of target cells when infected at low multiplicity of infection (MOI). Transcriptomics analysis identified selective alteration of mRNA processing and splicing modules that might underlie the potent anti-MM effects of combining HDACi and oHSV. In vivo, HDACi treatment increased intratumoral oHSV replication and boosted the capacity of oHSV to control the growth of human MM xenografts. Thus, our work supports further translational development of the combination approach employing HDACi and oHSV for the treatment of MM.
Collapse
Affiliation(s)
- Yoichiro Kawamura
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingyang Hua
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Alessandra Gurtner
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ego Wong
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Juri Kiyokawa
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nadia Shah
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Samuel D. Rabkin
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Robert L. Martuza
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Correspondence to: Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA. (H. Wakimoto)
| |
Collapse
|
12
|
Corniola MV, Meling TR. Management of Recurrent Meningiomas: State of the Art and Perspectives. Cancers (Basel) 2022; 14:cancers14163995. [PMID: 36010988 PMCID: PMC9406695 DOI: 10.3390/cancers14163995] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Intracranial meningiomas account for 30% to 40% of the primary lesions of the central nervous system. Surgery is the mainstay treatment whenever symptoms related to an intra-cranial meningioma are encountered. However, the management of recurrences after initial surgery, which are not uncommon, is still a matter of debate. Here, we present the alternatives described in the management of meningioma recurrence (radiotherapy, stereotaxic radiosurgery, protontherapy, and chemotherapy, among others). Their overall results are compared to surgery and future perspectives are presented. Abstract Background: While meningiomas often recur over time, the natural history of repeated recurrences and their management are not well described. Should recurrence occur, repeat surgery and/or use of adjuvant therapeutic options may be necessary. Here, we summarize current practice when it comes to meningioma recurrence after initial surgical management. Methods: A total of N = 89 articles were screened. N = 41 articles met the inclusion criteria and N = 16 articles failed to assess management of meningioma recurrence. Finally, N = 24 articles were included in our review. Results: The articles were distributed as follows: studies on chemotherapy (N = 14), radiotherapy, protontherapy, and stereotaxic radiosurgery (N = 6), boron-neutron capture therapy (N = 2) and surgery (N = 3). No study seems to provide serious alternatives to surgery in terms of progression-free and overall survival. Recurrence can occur long after the initial surgery and also affects WHO grade 1 meningiomas, even after initial gross total resection at first surgery, emphasizing the need for a long-term and comprehensive follow-up. Conclusions: Surgery still seems to be the state-of-the-art management when it comes to meningioma recurrence, since none of the non-surgical alternatives show promising results in terms of progression-free and overall survival.
Collapse
Affiliation(s)
- Marco Vincenzo Corniola
- Service de Neurochirurgie, Pôle des Neurosciences, Centre Hospitalier Universitaire de Rennes, 35000 Rennes, France
- Faculté de Médecine, Université de Rennes 1, 35000 Rennes, France
- Faculté de Médecine, Université de Genève, 1205 Geneve, Switzerland
- Laboratoire du Traitement de Signal, Unité Médicis, INSERM UMR 1099 LTSI, Université de Rennes 1, 35000 Rennes, France
| | - Torstein R. Meling
- Faculté de Médecine, Université de Genève, 1205 Geneve, Switzerland
- Department of Neurosurgery, Geneva University Hospitals, 1205 Geneva, Switzerland
- Besta NeuroSim Center, Fondazione IRCCS, Istituto Neurologico Carlo Basta, 20133 Milano, Italy
- Correspondence:
| |
Collapse
|
13
|
王 瀚, 王 跃, 刘 志, 徐 建. [Targeted Drug Therapy for Intracranial Tumors]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2022; 53:564-572. [PMID: 35871724 PMCID: PMC10409465 DOI: 10.12182/20220760102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Indexed: 06/15/2023]
Abstract
Intracranial tumors seriously affect the physical and mental health of humans. Due to variations in the nature and the growth site of tumors, individualized and specific treatment of patients with intracranial tumor has become a hotspot topic of research, and targeted drug therapy of intracranial tumors, an important subspecialty of precision medicine, has become a key issue that scientists are working hard to tackle. At present, the rapid development in molecular biology and genomics has provided corresponding targets for precision therapies of tumors. However, the blood-brain barrier and blood-tumor barrier prevent drugs from reaching intracranial targets. Therefore, finding effective ways to elevate the concentration of intracranial drugs has become the key issue concerning existing targeted therapies for intracranial tumors. Herein, we reviewed the current status of targeted drug therapy for different intracranial tumors and discussed their efficacy, intending to provide new perspectives for the treatment of intracranial tumors with targeted drugs in the future.
Collapse
Affiliation(s)
- 瀚 王
- 四川大学华西医院 神经外科 (成都 610041)Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
- 宜宾市第二人民医院•四川大学华西医院宜宾医院 神经外科 (宜宾 644000)Department of Neurosurgery, the Second People's Hospital of Yibin, West China Hospital, Sichuan University, Yibin 644000, China
| | - 跃龙 王
- 四川大学华西医院 神经外科 (成都 610041)Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 志勇 刘
- 四川大学华西医院 神经外科 (成都 610041)Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 建国 徐
- 四川大学华西医院 神经外科 (成都 610041)Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
14
|
Graillon T, Tabouret E, Chinot O. Chemotherapy and targeted therapies for meningiomas: what is the evidence? Curr Opin Neurol 2021; 34:857-867. [PMID: 34629433 DOI: 10.1097/wco.0000000000001002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Although most meningiomas are slow growing tumors mainly controlled by surgery with or without radiotherapy, aggressive meningiomas that fail these conventional treatments constitute a rare situation, a therapeutic challenge and an unmet need in neuro-oncology. RECENT FINDING Mutational landscape in recurrent high-grade meningiomas includes mainly NF2 mutation or 22q chromosomal deletion, whereas telomerase reverse transcriptase promoter, BAP-1 and CDK2NA mutations were also found in aggressive meningiomas. Pi3K-Akt-mTOR pathway is currently the most relevant intracellular signaling pathway target in meningiomas with preliminary clinical activity observed. Assessment of drug activity with progression free survival rate at 6 months is challenging in regard to meningioma growth rate heterogeneity, so that 3-dimensional growth rate before and during treatment could be considered in the future to selected new active drugs. SUMMARY Despite a low evidence level, some systemic therapies may be considered for patients with recurrent meningioma not amenable to further surgery or radiotherapy. In recurrent high-grade meningioma, everolimus-octreotide combination, bevacizumab, sunitinib and peptide receptor radionuclide therapy exhibit a signal of activity that may justify their clinical use. Despite a lack of clear signal of activity to date, immunotherapy may offer new perspectives in the treatment of these refractory tumors.
Collapse
Affiliation(s)
- Thomas Graillon
- Aix Marseille Univ, APHM, INSERM, MMG, UMR1251, La Timone Hospital, neurosurgery department Marseille, France
| | - Emeline Tabouret
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, La Timone Hospital, Neurooncology Department, Marseille, France
| | - Olivier Chinot
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, La Timone Hospital, Neurooncology Department, Marseille, France
| |
Collapse
|
15
|
Tonogai EJ, Huang S, Botham RC, Berry MR, Joslyn SK, Daniel GB, Chen Z, Rao J, Zhang X, Basuli F, Rossmeisl JH, Riggins GJ, LeBlanc AK, Fan TM, Hergenrother PJ. Evaluation of a procaspase-3 activator with hydroxyurea or temozolomide against high-grade meningioma in cell culture and canine cancer patients. Neuro Oncol 2021; 23:1723-1735. [PMID: 34216463 PMCID: PMC8485451 DOI: 10.1093/neuonc/noab161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND High-grade meningioma is an aggressive type of brain cancer that is often recalcitrant to surgery and radiotherapy, leading to poor overall survival. Currently, there are no FDA-approved drugs for meningioma, highlighting the need for new therapeutic options, but development is challenging due to the lack of predictive preclinical models. METHODS To leverage the known overexpression of procaspase-3 in meningioma, PAC-1, a blood-brain barrier penetrant procaspase-3 activator, was evaluated for its ability to induce apoptosis in meningioma cells. To enhance the effects of PAC-1, combinations with either hydroxyurea or temozolomide were explored in cell culture. Both combinations were further investigated in small groups of canine meningioma patients and assessed by MRI, and the novel apoptosis tracer, [18F]C-SNAT4, was evaluated in patients treated with PAC-1 + HU. RESULTS In meningioma cell lines in culture, PAC-1 + HU are synergistic while PAC-1 + TMZ show additive-to-synergistic effects. In canine meningioma patients, PAC-1 + HU led to stabilization of disease and no change in apoptosis within the tumor, whereas PAC-1 + TMZ reduced tumor burden in all three canine patients treated. CONCLUSIONS Our results suggest PAC-1 + TMZ as a potentially efficacious combination for the treatment of human meningioma, and also demonstrate the utility of including pet dogs with meningioma as a means to assess anticancer strategies for this common brain tumor.
Collapse
Affiliation(s)
- Emily J Tonogai
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Shan Huang
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Rachel C Botham
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Matthew R Berry
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | | | - Gregory B Daniel
- Radiology, Department of Small Animal Clinical Sciences, Virgina-Maryland College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - Zixin Chen
- Departments of Radiology and Chemistry, Stanford Medicine, Stanford, California, USA
| | - Jianghong Rao
- Departments of Radiology and Chemistry, Stanford Medicine, Stanford, California, USA
| | - Xiang Zhang
- Chemistry and Synthesis Center, NHLBI, NIH, Bethesda, Maryland, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, NHLBI, NIH, Bethesda, Maryland, USA
| | - John H Rossmeisl
- Neurology and Neurosurgery, Department of Small Animal Clinical Sciences, Virgina-Maryland College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - Gregory J Riggins
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Amy K LeBlanc
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Timothy M Fan
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
16
|
Brain-invasive meningiomas: molecular mechanisms and potential therapeutic options. Brain Tumor Pathol 2021; 38:156-172. [PMID: 33903981 DOI: 10.1007/s10014-021-00399-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
Meningiomas are the most commonly diagnosed benign intracranial adult tumors. Subsets of meningiomas that present with extensive invasion into surrounding brain areas have high recurrence rates, resulting in difficulties for complete resection, substantially increased mortality of patients, and are therapeutically challenging for neurosurgeons. Exciting new data have provided insights into the understanding of the molecular machinery of invasion. Moreover, clinical trials for several novel approaches have been launched. Here, we will highlight the mechanisms which govern brain invasion and new promising therapeutic approaches for brain-invasive meningiomas, including pharmacological approaches targeting three major aspects of tumor cell invasion: extracellular matrix degradation, cell adhesion, and growth factors, as well as other innovative treatments such as immunotherapy, hormone therapy, Tumor Treating Fields, and biodegradable copolymers (wafers), impregnated chemotherapy. Those ongoing studies can offer more diversified possibilities of potential treatments for brain-invasive meningiomas, and help to increase the survival benefits for patients.
Collapse
|
17
|
Abstract
Meningiomas are the most frequently occurring primary brain tumors in adults, representing almost one-third of all primary central nervous system tumors. Several factors have been suggested as an underlying cause in the development of meningiomas, such as ionizing radiation (therapeutic or other incidental exposure), hormonal factors, and genetic predisposition syndromes. Other established factors associated with meningiomas include age, female gender, and those from non-Hispanic Black backgrounds. Though the 2016 World Health Organization Classification of Brain Tumors largely preserves the existing grading scheme for organization of meningioma, there is increasing understanding of the molecular factors underlying the development of meningioma, some of which now form the basis for active clinical investigation. The mainstay of treatment has been the combination of radiation therapy and surgery, with a limited role for systemic therapy due to low efficacy, short duration of treatment response, and lack of uniform response criteria. Similar to other primary and metastatic brain tumors, immune-based therapies hold promise and are still under investigation.
Collapse
Affiliation(s)
- Ugonma N Chukwueke
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, MA, United States.
| |
Collapse
|
18
|
|
19
|
Wilson TA, Huang L, Ramanathan D, Lopez-Gonzalez M, Pillai P, De Los Reyes K, Kumal M, Boling W. Review of Atypical and Anaplastic Meningiomas: Classification, Molecular Biology, and Management. Front Oncol 2020; 10:565582. [PMID: 33330036 PMCID: PMC7714950 DOI: 10.3389/fonc.2020.565582] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Although the majority of meningiomas are slow-growing and benign, atypical and anaplastic meningiomas behave aggressively with a penchant for recurrence. Standard of care includes surgical resection followed by adjuvant radiation in anaplastic and partially resected atypical meningiomas; however, the role of adjuvant radiation for incompletely resected atypical meningiomas remains debated. Despite maximum treatment, atypical, and anaplastic meningiomas have a strong proclivity for recurrence. Accumulating mutations over time, recurrent tumors behave more aggressively and often become refractory or no longer amenable to further surgical resection or radiation. Chemotherapy and other medical therapies are available as salvage treatment once standard options are exhausted; however, efficacy of these agents remains limited. This review discusses the risk factors, classification, and molecular biology of meningiomas as well as the current management strategies, novel therapeutic approaches, and future directions for managing atypical and anaplastic meningiomas.
Collapse
Affiliation(s)
| | - Lei Huang
- Loma Linda University, Loma Linda, CA, United States
| | | | | | - Promod Pillai
- Loma Linda University, Loma Linda, CA, United States
| | | | | | - Warren Boling
- Loma Linda University, Loma Linda, CA, United States
| |
Collapse
|
20
|
Garzon-Muvdi T, Bailey DD, Pernik MN, Pan E. Basis for Immunotherapy for Treatment of Meningiomas. Front Neurol 2020; 11:945. [PMID: 32982948 PMCID: PMC7483661 DOI: 10.3389/fneur.2020.00945] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Meningiomas are common tumors that account for approximately one third of CNS tumors diagnosed every year. They are classified by the World Health Organization in grades I-III. Higher grades have an increased rate of growth, invasiveness, rate of recurrence, and worse outcomes than lower grades. Most meningiomas are grade I, while ~18% of meningiomas are grade II and III in hospital-based series. Meningiomas are typically "benign" tumors that are treated with surgery and radiation. However, when they recur or are unresectable, treatment options are very limited, especially since they are chemotherapy-resistant. Recent advances in the treatment of cancers with immunotherapy have focused on checkpoint blockade as well as other types of immunotherapy. There is emerging evidence supporting the use of immunotherapy as a potentially effective treatment strategy for meningioma patients. The immune microenvironment of meningiomas is a complex interplay of genetic alterations, immunomodulatory protein expression, and tumor-immune cell interactions. Meningiomas are known to be infiltrated by immune cells including microglia, macrophages, B-cells, and T-cells. Several mechanisms contribute to decreased an ti-tumor immune response, allowing tumor growth and evasion of the immune system. We discuss the most current knowledge on the immune micro-environment of meningiomas, preclinical findings of immunotherapy in meningiomas, meningioma immunotherapy clinical trials, and also offer insight into future prospects for immunotherapies in meningiomas.
Collapse
Affiliation(s)
- Tomas Garzon-Muvdi
- Department of Neurosurgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Destiny D. Bailey
- Department of Neurosurgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Mark N. Pernik
- Department of Neurosurgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Edward Pan
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
21
|
Zhao L, Zhao W, Hou Y, Wen C, Wang J, Wu P, Guo Z. An Overview of Managements in Meningiomas. Front Oncol 2020; 10:1523. [PMID: 32974188 PMCID: PMC7473392 DOI: 10.3389/fonc.2020.01523] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/16/2020] [Indexed: 12/16/2022] Open
Abstract
Meningioma is the most frequent primary tumor of the central nervous system. Important advances have been achieved in the treatment of meningioma in recent decades. Although most meningiomas are benign and have a good prognosis after surgery, clinicians often face challenges when the morphology of the tumor is complicated or the tumor is close to vital brain structures. At present, the longstanding treatment strategies of meningioma are mainly surgery and radiotherapy. The effectiveness of systemic therapy, such as chemotherapy or targeted therapy, has not been confirmed by big data series, and some clinical trials are still in progress. In this review, we summarize current treatment strategies and future research directions for meningiomas.
Collapse
Affiliation(s)
- Lianhua Zhao
- Department of Neurology, Tianjin TEDA Hospital, Tianjin, China
| | - Wei Zhao
- Department of Neurology, Tianjin TEDA Hospital, Tianjin, China
| | - Yanwei Hou
- Department of Neurosurgery, Tianjin TEDA Hospital, Tianjin, China
| | - Cuixia Wen
- Department of Radiotherapy, Xuzhou Central Hospital, Xuzhou, China
| | - Jing Wang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Pei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zaiyu Guo
- Department of Neurosurgery, Tianjin TEDA Hospital, Tianjin, China
| |
Collapse
|
22
|
Hrachova M, Nguyen ENT, Fu BD, Dandekar MJ, Kong XT, Cadena G, Hsu FPK, Billimek J, Taylor TH, Bota DA. A Retrospective Interventional Cohort Study to Assess the Safety and Efficacy of Sandostatin LAR for Treatment of Recurrent and/or Refractory Meningiomas. Front Neurol 2020; 11:373. [PMID: 32435228 PMCID: PMC7218113 DOI: 10.3389/fneur.2020.00373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Meningiomas are the most common adult primary intracranial tumors in the United States. Despite high recurrence rate of atypical and malignant subtypes, there is no approved drug indicated specifically for meningioma. Since the majority of meningiomas exhibit high density of somatostatin receptors subtypes, somatostatin analogs have been under close investigation. The aim of this study was to evaluate efficacy and safety of Sandostatin LAR (octreotide) in patients with progressive, and/or recurrent meningioma, and identify subset of patients who were more likely to benefit from this treatment. Methods: A total of 43 patients ≥ 18 years old were included in the retrospective chart review. The patients underwent treatment with Sandostatin LAR (octreotide) from 01.01.2010 to 06.01.2017 at the University of California, Irvine after confirmation of the diagnosis. Six months progression free survival (PFS6) was defined as a primary endpoint, and the overall survival (OS), safety, and toxicity were identified as secondary endpoints. Results: The OS for 6 months, 1, and 3 years for all WHO grades was 94.8, 88.1, and 67.0%, respectively. The PFS6 for WHO I, II, III, and all was 89.4, 89, 33.3, and 80% respectively. For patients with no prior surgeries, chemotherapy or radiation, the PFS6 was 88.9, 84.8, and 94.8%, respectively. Interestingly, the PFS6 was 90.5% for skull-based and 80% for 3–6 cm tumors. Patients with tumors in parasagittal location had PFS6 of 83.3% compared to PFS6 of 50.0% for patients with convexity tumors. Evaluation of PFS6 based on the effect of estrogen and progesterone on meningioma identified that ER-PR+ tumors had PFS6 of 87.8% while patients with ER-PR- meningiomas had PFS6 of 62.5%. Median TTP for WHO grade I, II, and III was 3.1, 2.40, and 0.26 years, respectively. Subgroup analysis showed that median TTP was 3.1 years for <3 cm tumors, 3.22 years for skull-based tumors, 2.37 years for patients with prior surgeries and 3.10 years for patients with no history of chemotherapy. History of radiation had no effect on median TTP. Sandostatin LAR (octreotide) was well-tolerated. Conclusions:This is one of the largest retrospective analysis of meningioma patients treated with Sandostatin LAR (octreotide) suggesting that this treatment has minimal to no adverse events and could prolong overall survival, and progression free survival especially for patients with ER-PR+ tumors who underwent surgeries for small skull-based tumors.
Collapse
Affiliation(s)
- Maya Hrachova
- Department of Neurology, Irvine Medical Center, University of California, Orange, Orange, CA, United States
| | - Emely Nhi T Nguyen
- School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Beverly D Fu
- Department of Neurology, Irvine Medical Center, University of California, Orange, Orange, CA, United States.,Chao Family Comprehensive Cancer Center, Irvine Medical Center, University of California, Orange, Orange, CA, United States
| | - Manisha J Dandekar
- Chao Family Comprehensive Cancer Center, Irvine Medical Center, University of California, Orange, Orange, CA, United States
| | - Xiao-Tang Kong
- Department of Neurology, Irvine Medical Center, University of California, Orange, Orange, CA, United States.,Chao Family Comprehensive Cancer Center, Irvine Medical Center, University of California, Orange, Orange, CA, United States.,Department of Neurological Surgery, Irvine Medical Center, University of California, Orange, Orange, CA, United States
| | - Gilbert Cadena
- Chao Family Comprehensive Cancer Center, Irvine Medical Center, University of California, Orange, Orange, CA, United States.,Department of Neurological Surgery, Irvine Medical Center, University of California, Orange, Orange, CA, United States
| | - Frank P K Hsu
- Chao Family Comprehensive Cancer Center, Irvine Medical Center, University of California, Orange, Orange, CA, United States.,Department of Neurological Surgery, Irvine Medical Center, University of California, Orange, Orange, CA, United States
| | - John Billimek
- School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Thomas H Taylor
- Department of Epidemiology, University of California, Irvine, Irvine, CA, United States
| | - Daniela A Bota
- Department of Neurology, Irvine Medical Center, University of California, Orange, Orange, CA, United States.,School of Medicine, University of California, Irvine, Irvine, CA, United States.,Chao Family Comprehensive Cancer Center, Irvine Medical Center, University of California, Orange, Orange, CA, United States.,Department of Neurological Surgery, Irvine Medical Center, University of California, Orange, Orange, CA, United States
| |
Collapse
|
23
|
Medina-Lopes MDD, Augusto Casulari L. Treatment of a Woman with Inoperable Meningioma Using Mifepristone for 26 Years. Case Rep Neurol Med 2020; 2020:5162918. [PMID: 32095298 PMCID: PMC7036131 DOI: 10.1155/2020/5162918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/28/2019] [Accepted: 12/23/2019] [Indexed: 11/17/2022] Open
Abstract
Meningioma treatment includes observation of its growth or surgery with or without associated radiotherapy. However, drug treatment can be used for tumors deemed inoperable because of their size and location. Due to the presence of progesterone receptors, the use of antiprogestin mifepristone is recommended. This study describes a case of inoperable meningioma treated with mifepristone for 26 years without interruption. The patient is a 45-year-old woman diagnosed with plaque meningioma, extending from the bottom of her right orbit, through the length of the small wing of the sphenoid, part of the large wing of the sphenoid, especially near the superior orbital fissure, and at the ceiling of the orbit. As this meningioma was considered inoperable, treatment with 200 mg oral mifepristone was administered uninterruptedly for 26 years. This treatment initially halted the growth of the meningioma and subsequently resulted in a small reduction of its volume; however, the meningioma has persisted until the last evaluation. After five years of mifepristone use, hydroxyurea was added for nine months but was discontinued due to anemia and leucopenia. In conclusion, mifepristone was useful for the survival of the patient for those 26 years. The drug interfered with the natural history of the meningioma, which generally evolves to death in such long follow-up durations without associated surgery or radiation therapy.
Collapse
Affiliation(s)
| | - Luiz Augusto Casulari
- The University Hospital of Brasília and Clinic of Endocrinology and Neurology (CLINEN), Brasília, DF, Brazil
| |
Collapse
|
24
|
Abstract
Surgery is curative for most meningiomas, but a minority of these tumors recur and progress after resection. Initial trials of medical therapies for meningioma utilized nonspecific cytotoxic chemotherapies. The presence of hormone receptors on meningioma ushered in trials of hormone-mimicking agents. While these trials expanded clinical understanding of meningioma, they ultimately had limited efficacy in managing aggressive lesions. Subsequent detection of misregulated proteins and genomic aberrancies motivated the study of therapies targeting specific biological disturbances observed in meningioma. These advances led to trials of targeted kinase inhibitors and immunotherapies, as well as combinations of these agents together with chemotherapies. Prospective trials currently recruiting participants are testing a diverse range of medical therapies for meningioma, and some studies now require the presence of a specific protein alteration or genetic mutation as an inclusion criterion. Increasing understanding of the unique and heterogeneous nature of meningiomas will continue to spur the development of novel medical therapies for the arsenal against aggressive tumors.
Collapse
|
25
|
|
26
|
Nigim F, Wakimoto H, Kasper EM, Ackermans L, Temel Y. Emerging Medical Treatments for Meningioma in the Molecular Era. Biomedicines 2018; 6:biomedicines6030086. [PMID: 30082628 PMCID: PMC6165537 DOI: 10.3390/biomedicines6030086] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/31/2018] [Indexed: 12/19/2022] Open
Abstract
Meningiomas are the most common type of primary central nervous system tumors. Approximately, 80% of meningiomas are classified by the World Health Organization (WHO) as grade I, and 20% of these tumors are grade II and III, considered high-grade meningiomas (HGMs). Clinical control of HGMs, as well as meningiomas that relapse after surgery, and radiation therapy is difficult, and novel therapeutic approaches are necessary. However, traditional chemotherapies, interferons, hormonal therapies, and other targeted therapies have so far failed to provide clinical benefit. During the last several years, next generation sequencing has dissected the genetic heterogeneity of meningioma and enriched our knowledge about distinct oncogenic pathways driving different subtypes of meningiomas, opening up a door to new personalized targeted therapies. Molecular classification of meningioma allows a new design of clinical trials that assign patients to corresponding targeted agents based on the tumor genetic subtypes. In this review, we will shed light on emerging medical treatments of meningiomas with a particular focus on the new targets identified with genomic sequencing that have led to clinical trials testing novel compounds. Moreover, we present recent development of patient-derived preclinical models that provide platforms for assessing targeted therapies as well as strategies with novel mechanism of action such as oncolytic viruses.
Collapse
Affiliation(s)
- Fares Nigim
- Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Hiroaki Wakimoto
- Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Ekkehard M Kasper
- Department of Neurosurgery, McMaster University, Hamilton, ON 8L8 2X2, Canada.
| | - Linda Ackermans
- Department of Neurosurgery and Neuroscience, Maastricht University Medical Center, 6229 HY Maastricht, The Netherlands.
| | - Yasin Temel
- Department of Neurosurgery and Neuroscience, Maastricht University Medical Center, 6229 HY Maastricht, The Netherlands.
| |
Collapse
|
27
|
Franke AJ, Skelton IV WP, Woody LE, Bregy A, Shah AH, Vakharia K, Komotar RJ. Role of bevacizumab for treatment-refractory meningiomas: A systematic analysis and literature review. Surg Neurol Int 2018; 9:133. [PMID: 30090665 PMCID: PMC6057170 DOI: 10.4103/sni.sni_264_17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 05/22/2018] [Indexed: 01/17/2023] Open
Abstract
Background Meningiomas are the most prevalent primary tumor of the central nervous system (CNS), and although the majority of these neoplasms are classified as benign, nearly one fourth of the lesions display an aggressive profile characterized by pleomorphic histology, high recurrence rates, and overall resistance to standard treatment. Despite the ubiquitous nature of these tumors, no adjuvant therapeutic regimen has been identified which effectively controls disease recurrence and progression after surgery and radiation, leading to a dismal prognosis in this patient population. The primary focus of this research study is, hence, to assess the recently emerging use of bevacizumab, an anti-angiogenic agent, in the treatment of meningiomas. This systematic literature review analyzes the efficacy and safety of therapeutic bevacizumab for treatment-refractory meningiomas. Methods A systematic PubMed search was conducted according to PRISMA guidelines to identify all relevant reports investigating the anti-angiogenic agent bevacizumab in the treatment of intracranial meningiomas. The reported parameters from pertinent retrospective reviews, prospective studies, and case studies were volumetric reduction, radiographic response, clinical stability, overall survival (OS), and progression free survival (PFS) measured at 6 and 12 months postinitiation of treatment. Complications were cataloged based on the range and severity of the therapy-related toxicities. Results A total of 11 articles, 5 retrospective series, 2 prospective trials, and 4 case reports, reporting on a total of 92 patients, were included in this review. The use of bevacizumab therapy for intracranial meningiomas demonstrated median overall PFS of 16.8 months (range: 6.5-22 months) and PFS-6 of 73% (range: 44%-93%). Conclusions Therapeutic bevacizumab, either alone or with combination chemotherapies, for select patient populations with recurrent or progressive meningiomas, offers a treatment option that confers improved overall progression-free survival. To assess OS parameters, larger randomized controlled trials assessing the use of anti-angiogenic agents for recurrent/progressive meningiomas are warranted.
Collapse
Affiliation(s)
- Aaron J. Franke
- Department of Internal Medicine, University of Florida College of Medicine, Gainesville, USA
| | - William Paul Skelton IV
- Department of Internal Medicine, University of Florida College of Medicine, Gainesville, USA
| | | | - Amade Bregy
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ashish H. Shah
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kunal Vakharia
- Department of Neurological Surgery, University at Buffalo School of Medicine, Buffalo, New York, USA
| | - Ricardo J. Komotar
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
28
|
Sharma P, Katiyar V, Sharma R, Gurjar HK, Krishnan S. Letter: Role of Tyrosine Kinase Inhibitors in Recurrent Meningiomas: Controversies and Promises. Neurosurgery 2018; 82:E181-E183. [DOI: 10.1093/neuros/nyy055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
29
|
Visceral and bone metastases of a WHO grade 2 meningioma: A case report and review of the literature. Cancer Radiother 2017; 21:55-59. [DOI: 10.1016/j.canrad.2016.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 09/11/2016] [Accepted: 09/16/2016] [Indexed: 11/23/2022]
|
30
|
Balasubramanian SK, Sharma M, Silva D, Karivedu V, Schmitt P, Stevens GH, Barnett GH, Prayson RA, Elson P, Suh JH, Murphy ES, Chao ST. Longitudinal experience with WHO Grade III (anaplastic) meningiomas at a single institution. J Neurooncol 2016; 131:555-563. [DOI: 10.1007/s11060-016-2321-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 11/08/2016] [Indexed: 11/28/2022]
|
31
|
Review of controversies in management of non-benign meningioma. J Clin Neurosci 2016; 31:37-46. [DOI: 10.1016/j.jocn.2016.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/21/2016] [Accepted: 03/27/2016] [Indexed: 11/23/2022]
|
32
|
Elmaci İ, Altinoz MA, Sav A, Yazici Z, Ozpinar A. Giving another chance to mifepristone in pharmacotherapy for aggressive meningiomas—A likely synergism with hydroxyurea? Curr Probl Cancer 2016; 40:229-243. [DOI: 10.1016/j.currproblcancer.2016.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/10/2016] [Accepted: 05/02/2016] [Indexed: 12/31/2022]
|
33
|
Abstract
INTRODUCTION Meningioma comprise 20-30% of all primary brain tumors. Notwithstanding surgery and radiotherapy, a subset of patients will manifest recurrent meningioma. Systemic therapy is recommended only when further surgery and radiotherapy are not possible. No prospective study with a high level of evidence is available to inform as to recommendations regarding systemic therapy. AREAS COVERED We aim to summarize systemic therapies for recurrent meningioma. Expert commentary: Hydroxurea, temozolomide, irinotecan, the combination of cyclophosphamide/adriamycine/vincristine, interferon-alpha, somatostatin analogs, mifepristone, megestrol acetate, imatinib, erlotinib and gefitinib are considered as having limited efficacy. Potential activity of VEGF (vascular endothelial growth factor) inhibitors such as sunitinib, valatinib, and bevacizumab is suggested in small non-controlled studies and requires validation in randomized trials. The identification of new prognostic markers such as TERT promoter mutations and potential new therapeutic targets, such as KLF4, AKT1, TRAF7, and SMO mutations hopefully facilitate this endeavor.
Collapse
Affiliation(s)
- E Le Rhun
- a Lille University, PRISM Inserm U1191 , Villeneuve d'Ascq , France.,b Neuro-oncology, Department of Neurosurgery , Lille Universisty Hospital , Lille Cedex , France.,c Breast unit, Department of Medical Oncology , Oscar Lambret Center , Lille Cedex , France
| | - S Taillibert
- d Department of Neurology Mazarin , Pitié-Salpétrière Hospital, Assistance Publique des Hôpitaux de Paris , Paris , France.,e Department of Neurology , University Pierre et Marie Curie, Paris VI , Paris , France
| | - M C Chamberlain
- f Department of Neurology and Neurological Surgery , University of Washington , Seattle , WA , USA
| |
Collapse
|
34
|
Seystahl K, Stoecklein V, Schüller U, Rushing E, Nicolas G, Schäfer N, Ilhan H, Pangalu A, Weller M, Tonn JC, Sommerauer M, Albert NL. Somatostatin receptor-targeted radionuclide therapy for progressive meningioma: benefit linked to 68Ga-DOTATATE/-TOC uptake. Neuro Oncol 2016; 18:1538-1547. [PMID: 27106404 DOI: 10.1093/neuonc/now060] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 03/16/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The prognosis of patients with progressive meningioma after failure of surgery and radiotherapy is poor. METHODS We retrospectively evaluated the safety and efficacy of somatostatin-receptor (SSTR)-targeted radionuclide therapy (177Lu-DOTATATE [n = 16], 90Y-DOTATOC [n = 3], or both [n = 1]) in patients with progressive, treatment-refractory meningiomas (5 World Health Organization [WHO] grade I, 7 WHO grade II, 8 WHO grade III) and in part multifocal disease (17 of 20 patients). RESULTS SSTR radionuclide treatment (median of 3 treatment cycles, median administered dose/cycle 7400 MBq) led to a disease stabilization in 10 of 20 patients for a median time of 17 months. Stratification according to WHO grade showed a median progression-free survival (PFS) of 32.2 months for grade I tumors, 7.2 for grade II, and 2.1 for grade III. PFS at 6 months was 100% for grade I, 57% for grade II, and 0% for grade III. Median overall survival was 17.2 months in WHO grade III patients and not reached for WHO I and II at a median follow-up of 20 months. In the analysis of single meningioma lesions, maximal and mean standardized uptake values in pretherapeutic 68Ga-DOTATOC/-TATE PET/CT were significantly higher in those lesions with radiographic stability after 6 months. In line with this, high expression of SSTR via immunohistochemistry was associated with PFS >6 months. CONCLUSIONS SSTR-targeted radionuclide treatment has activity in a subset of patients with meningioma. Expression of SSTR via immunohistochemistry or radionuclide uptake might serve as a predictive biomarker for outcome to facilitate individualized treatment optimization in patients with uni- and multifocal meningiomas.
Collapse
Affiliation(s)
- Katharina Seystahl
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Veit Stoecklein
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Ulrich Schüller
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Elisabeth Rushing
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Guillaume Nicolas
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Niklaus Schäfer
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Harun Ilhan
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Athina Pangalu
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Michael Weller
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Jörg-Christian Tonn
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Michael Sommerauer
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| | - Nathalie L Albert
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland (K.S., M.W., M.S.); Department of Neurosurgery, University Hospital LMU Munich, Munich, Germany (V.S., J.-C.T.); Department of Neuropathology, University Hospital LMU Munich, Munich, Germany (U.S.); Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland (E.R.); Department of Nuclear Medicine, University Hospital Basel, Basel, Switzerland (G.N.); Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (N.S., M.S.); Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany (H.I., N.L.A.); Department of Neuroradiology, University Hospital Zurich, Zurich, Switzerland (A.P.)
| |
Collapse
|
35
|
Karsy M, Guan J, Cohen A, Colman H, Jensen RL. Medical Management of Meningiomas: Current Status, Failed Treatments, and Promising Horizons. Neurosurg Clin N Am 2016; 27:249-60. [PMID: 27012389 DOI: 10.1016/j.nec.2015.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Meningiomas are benign tumors of the central nervous system, with low recurrence risk for World Health Organization (WHO) grade I lesions but a high risk for WHO grade II and III lesions. Current standard treatments include maximum safe surgical resection when indicated and radiation. Only three systemic therapies alpha-interferon, somatostatin receptor agonists, and vascular endothelial growth factor inhibitors are currently recommended by the National Comprehensive Cancer Network for treatment of recurrent meningioma. This paper aims to review medical approaches in the treatment of meningiomas.
Collapse
Affiliation(s)
- Michael Karsy
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, 175 N. Medical Drive East, Salt Lake City, UT 84132, USA
| | - Jian Guan
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, 175 N. Medical Drive East, Salt Lake City, UT 84132, USA
| | - Adam Cohen
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Howard Colman
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, 175 N. Medical Drive East, Salt Lake City, UT 84132, USA; Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Randy L Jensen
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, 175 N. Medical Drive East, Salt Lake City, UT 84132, USA; Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA; Department of Radiation Oncology, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA.
| |
Collapse
|
36
|
Karsy M, Hoang N, Barth T, Burt L, Dunson W, Gillespie DL, Jensen RL. Combined Hydroxyurea and Verapamil in the Clinical Treatment of Refractory Meningioma: Human and Orthotopic Xenograft Studies. World Neurosurg 2016; 86:210-9. [DOI: 10.1016/j.wneu.2015.09.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 09/16/2015] [Accepted: 09/19/2015] [Indexed: 11/26/2022]
|
37
|
Fogh SE, Johnson DR, Barker FG, Brastianos PK, Clarke JL, Kaufmann TJ, Oberndorfer S, Preusser M, Raghunathan A, Santagata S, Theodosopoulos PV. Case-Based Review: meningioma. Neurooncol Pract 2016; 3:120-134. [PMID: 31386096 DOI: 10.1093/nop/npv063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Indexed: 12/30/2022] Open
Abstract
Meningioma is by far the most common primary intracranial tumor in adults. Treatment of meningioma is complex due to a tremendous amount of variability in tumor behavior. Many patients are incidentally found to have tumors that will remain asymptomatic throughout their lives. It is important to identify these patients so that they can be spared from potentially morbid interventions. On the other end of the spectrum, high-grade meningiomas can behave very aggressively. When treatment is necessary, surgical resection is the cornerstone of meningioma therapy. Studies spanning decades have demonstrated that extent of resection correlates with prognosis. Radiation therapy, either in the form of external beam radiation therapy or stereotactic radiosurgery, represents another important therapeutic tool that can be used in place of or as a supplement to surgery. There are no chemotherapeutic agents of proven efficacy against meningioma, and chemotherapy treatment is generally reserved for patients who have exhausted surgical and radiotherapy options. Ongoing and future studies will help to answer unresolved questions such as the optimum use of radiation in resected WHO grade II meningiomas and the efficacy of additional chemotherapy agents.
Collapse
Affiliation(s)
- Shannon E Fogh
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Derek R Johnson
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Fred G Barker
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Priscilla K Brastianos
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Jennifer L Clarke
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Timothy J Kaufmann
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Stephan Oberndorfer
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Matthias Preusser
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Aditya Raghunathan
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Sandro Santagata
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| | - Philip V Theodosopoulos
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA (S.E.F.); Department of Radiology, Mayo Clinic, Rochester, MN, USA (D.R.J., T.J.K.); Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (F.G.B.); Division of Neuro-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA (P.K.B.); Department of Neurology and Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (J.L.C.); Department of Neurology, Karl Landsteiner University Clinic, St Pölten, Austria (S.O.); Department of Internal Medicine, Medical University, Vienna, CCC, Austria (M.P.); Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA (A.R.); Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA (S.S.); Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA (P.V.T.)
| |
Collapse
|
38
|
Abstract
Intracranial meningiomas are tumors arising from the covering cells of the arachnoid layer of the dura mater or from the intraventricular choroid plexus. While mostly benign tumors, they still represent a major challenge to neurosurgeons and other medical disciplines involved in their diagnostic and therapeutic management. Although this review intends to give some state-of-the-art information from the literature, it is mainly based on personal experiences since more than 30 years caring for more than 1500 meningioma patients and point to a few new strategies to further improve on patient outcome.Diagnostics are based on magnetic resonance imaging which shows the relationship between tumor and surrounding intracranial structures, particularly the brain but also the vasculature and to some extent the cranial nerves. Furthermore, it may suggest the grading of the tumor and is very helpful in the postoperative diagnosis of complications and later follow-up course.Surgery still is the main treatment with the aim to completely remove the tumor; also in cases of recurrence, other additional options include radiotherapy and radiosurgery for incompletely removed or recurrent meningiomas. Postoperative chemotherapy has not been shown to provide substantial benefit to the patient especially in highly malignant meningiomas.All therapy options should be intended to provide the patient with the best possible functional outcome. Patients' perspective is not always equivalent to surgeons' perspectives. Neuropsychological evaluation and additional guidance of patients harboring meningiomas have proven to be important in modern neurosurgical intracranial tumor treatment. Their help beyond neurosurgical care facilitates the patients to lead an independent postoperative life.
Collapse
Affiliation(s)
- H Maximilian Mehdorn
- Department of Neurosurgery, University Clinics of Schleswig-Holstein Campus Kiel, Arnold Heller Str 3 Hs 41, 24105, Kiel, Germany.
| |
Collapse
|
39
|
Hydroxyurea with or without imatinib in the treatment of recurrent or progressive meningiomas: a randomized phase II trial by Gruppo Italiano Cooperativo di Neuro-Oncologia (GICNO). Cancer Chemother Pharmacol 2015; 77:115-20. [PMID: 26659583 DOI: 10.1007/s00280-015-2927-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/17/2015] [Indexed: 10/22/2022]
Abstract
PURPOSE Hydroxyurea (HU) is among the most widely used salvage therapies in progressive meningiomas. Platelet-derived growth factor receptors are expressed in virtually all meningiomas. Imatinib sensitizes transformed cells to the cytotoxic effects of chemotherapeutic agents that interfere with DNA metabolism. The combination of HU with imatinib yielded intriguing results in recurrent malignant glioma. The current trial addressed the activity of this association against meningioma. METHODS Patients with recurrent or progressive WHO grade I-III meningioma, without therapeutic indication for surgery, radiotherapy, or stereotactic radiosurgery, aged 18-75 years, ECOG performance status 0-2, and not on enzyme-inducing anti-epileptic drugs were randomized to receive HU 500 mg BID ± imatinib 400 mg QD until progression, unacceptable toxicity, or patient's refusal. The primary endpoint was progression-free survival rate at 9 months (PFS-9). RESULTS Between September 2009 and February 2012, 15 patients were randomized to receive HU + imatinib (N = 7; Arm A) or HU alone (N = 8; Arm B). Afterward the trial was prematurely closed due to slow enrollment rate. PFS-9 (A/B) was 0/75%, and median PFS was 4/19.5 months. Median and 2-year overall survival (A/B) rates were: 6/27.5 months; 28.5/75%, respectively. Main G3-4 toxicities were: G3 neutropenia in 1/0, G4 headache in 1/1, and G3 vomiting in 1/0. CONCLUSION The conduction of a study in recurrent or progressive meningioma remains a challenge. Given the limited number of patients enrolled, no firm conclusions can be drawn about the combination of imatinib and HU. The optimal systemic therapy for meningioma failing surgery and radiation has yet to be identified.
Collapse
|
40
|
Improvement in Visual Fields After Treatment of Intracranial Meningioma With Bevacizumab. J Neuroophthalmol 2015; 35:382-6. [DOI: 10.1097/wno.0000000000000253] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Ji Y, Rankin C, Grunberg S, Sherrod AE, Ahmadi J, Townsend JJ, Feun LG, Fredericks RK, Russell CA, Kabbinavar FF, Stelzer KJ, Schott A, Verschraegen C. Double-Blind Phase III Randomized Trial of the Antiprogestin Agent Mifepristone in the Treatment of Unresectable Meningioma: SWOG S9005. J Clin Oncol 2015; 33:4093-8. [PMID: 26527781 DOI: 10.1200/jco.2015.61.6490] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Progesterone receptors are expressed in approximately 70% of meningiomas. Mifepristone is an oral antiprogestational agent reported to have modest activity in a phase II study. This multicenter, prospective, randomized, placebo-controlled phase III trial conducted by SWOG was planned to define the role of mifepristone in the treatment of unresectable meningioma. PATIENTS AND METHODS Eligible patients were randomly assigned to receive either mifepristone or placebo for 2 years unless disease progressed. Patients who were stable or responding to protocol therapy after 2 years had the option to continue with the same blinded therapy. Serial follow-up allowed assessment of efficacy and toxicity. Time to treatment failure and overall survival were ascertained for all randomly assigned patients. On progression, patients receiving placebo could cross over and receive active drug. RESULTS Among 164 eligible patients, 80 were randomly assigned to mifepristone and 84 to placebo. Twenty-four patients (30%) were able to complete 2 years of mifepristone without disease progression, adverse effects, or other reasons for discontinuation. Twenty-eight patients (33%) in the placebo arm completed the 2-year study. There was no statistical difference between the arms in terms of failure-free or overall survival. CONCLUSION Long-term administration of mifepristone was well tolerated but had no impact on patients with unresectable meningioma.
Collapse
Affiliation(s)
- Yongli Ji
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Cathryn Rankin
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Steven Grunberg
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Andy E Sherrod
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Jamshid Ahmadi
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Jeannette J Townsend
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Lynn G Feun
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Ruth K Fredericks
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Christy A Russell
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Fairooz F Kabbinavar
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Keith J Stelzer
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Anne Schott
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI
| | - Claire Verschraegen
- Yongli Ji, Steven Grunberg, and Claire Verschraegen, University of Vermont Cancer Center, Burlington, VT; Cathryn Rankin, SWOG Statistical Center, Seattle, WA; Andy E. Sherrod, Jamshid Ahmadi, and Christy A. Russell, University of Southern California; Fairooz F. Kabbinavar, University of California Los Angeles, Los Angeles, CA; Jeannette J. Townsend, University of Utah Medical Center, Salt Lake City, UT; Lynn G. Feun, University of Miami, Miami, FL; Ruth K. Fredericks, University of Mississippi Medical Center, Jackson, MS; Keith J. Stelzer, Celilo Cancer Center, the Dalles, OR; and Anne Schott, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
42
|
Abstract
The efficacy of surgery and radiation has been well validated in the treatment of meningiomas, with efficacy depending on tumor pathology, size, symptomatology and rate of progression. The role of medical therapy has the least amount of data but is being increasingly investigated for tumors that are inoperable or those tumors that recur and/or progress despite standard therapy. In this review, current data on the use of chemotherapeutic agents in the management of meningiomas will be reviewed, including cytotoxic, biologic, targeted molecular and hormonal agents.
Collapse
Affiliation(s)
- Wendy J Sherman
- Northwestern University Department of Neurology, 710 North Lake Shore Drive, Abbott Hall, Room 1123, Chicago, IL 60611, USA
| | | |
Collapse
|
43
|
Sun SQ, Hawasli AH, Huang J, Chicoine MR, Kim AH. An evidence-based treatment algorithm for the management of WHO Grade II and III meningiomas. Neurosurg Focus 2015; 38:E3. [DOI: 10.3171/2015.1.focus14757] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The management of WHO Grade II “atypical” meningiomas (AMs) and Grade III “malignant” meningiomas (MMs) remains controversial and under-investigated in prospective studies. The roles of surgery, radiation therapy, radiosurgery, and chemotherapy have been incompletely delineated. This has left physicians to decipher how they should treat patients on a case-by-case basis. In this study, the authors review the English-language literature on the management and clinical outcomes associated with AMs and MMs diagnosed using the WHO 2000/2007 grading criteria. Twenty-two studies for AMs and 7 studies for MMs were examined in detail. The authors examined clinical decision points using the literature and concepts from evidence-based medicine. Acknowledging the retrospective nature of the studies concerning AM and MM, the authors did find evidence for the following clinical strategies: 1) maximal safe resection of AM and MM; 2) active surveillance after gross-total resection of AM; 3) adjuvant radiation therapy after subtotal resection of AM, especially in the absence of putative radioresistant features; and 4) adjuvant radiation therapy after resection of MM.
Collapse
Affiliation(s)
- Sam Q. Sun
- 1Washington University School of Medicine; and
| | | | - Jiayi Huang
- 3Radiation Oncology, Washington University School of Medicine in St. Louis, Missouri
| | | | | |
Collapse
|
44
|
Jung HW, Lee HC, Kim JH, Jang HM, Moon JH, Sur JH, Ha J, Jung DI. Imatinib mesylate plus hydroxyurea chemotherapy for cerebellar meningioma in a Belgian Malinois dog. J Vet Med Sci 2014; 76:1545-8. [PMID: 25131949 PMCID: PMC4272992 DOI: 10.1292/jvms.14-0001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An 8-year-old intact male Belgian Malinois, weighing 37.2 kg, was referred for evaluation due to right side facial paresis, ataxia and a 2-month history of decreased cognitive ability. Physical and neurological examinations revealed mild depression, left-sided head tilt, right-sided facial paresis and ataxia. A well-demarcated, broad-based cerebellar mass and hyperostosis were found on CT imaging of the brain. Based on these CT findings, a cerebellar meningioma was strongly suspected. Hydroxyurea and prednisolone were administered; after 4 weeks, there was reduction in mass size as compared to initial CT results. However, the mass size was found to have grown 6 weeks after hydroxyurea treatment. We then prescribed a combination of imatinib mesylate and hydroxyurea. Two weeks following combination treatment, the mass size had reduced significantly. The mass continuously decreased in size until the patient died during anesthesia. Cerebellar transitional meningioma was confirmed by histopathologic examination. To the author's knowledge, this is the first reported case of imatinib mesylate plus hydroxyurea therapy for the treatment of meningioma in veterinary medicine.
Collapse
Affiliation(s)
- Hae-Won Jung
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 660-701, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Kaley TJ, Wen P, Schiff D, Ligon K, Haidar S, Karimi S, Lassman AB, Nolan CP, DeAngelis LM, Gavrilovic I, Norden A, Drappatz J, Lee EQ, Purow B, Plotkin SR, Batchelor T, Abrey LE, Omuro A. Phase II trial of sunitinib for recurrent and progressive atypical and anaplastic meningioma. Neuro Oncol 2014; 17:116-21. [PMID: 25100872 DOI: 10.1093/neuonc/nou148] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND No proven effective medical therapy for surgery and radiation-refractory meningiomas exists. Sunitinib malate (SU011248) is a small-molecule tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor, abundant in meningiomas. METHODS This was a prospective, multicenter, investigator-initiated single-arm phase II trial. The primary cohort enrolled patients with surgery and radiation-refractory recurrent World Health Organization (WHO) grades II-III meningioma. An exploratory cohort enrolled patients with WHO grade I meningioma, hemangiopericytoma, or hemangioblastoma. Sunitinib was administered at 50 mg/d for days 1-28 of every 42-day cycle. The primary endpoint was the rate of 6-month progression-free survival (PFS6), with secondary endpoints of radiographic response rate, safety, PFS, and overall survival. Exploratory objectives include analysis of tumoral molecular markers and MR perfusion imaging. RESULTS Thirty-six patients with high-grade meningioma (30 atypical and 6 anaplastic) were enrolled. Patients were heavily pretreated (median number of 5 recurrences, range 2-10). PFS6 rate was 42%, meeting the primary endpoint. Median PFS was 5.2 months (95% CI: 2.8-8.3 mo), and median overall survival was 24.6 months (95% CI: 16.5-38.4 mo). Thirteen patients enrolled in the exploratory cohort. Overall toxicity included 1 grade 5 intratumoral hemorrhage, 2 grade 3 and 1 grade 4 CNS/intratumoral hemorrhages, 1 grade 3 and 1 grade 4 thrombotic microangiopathy, and 1 grade 3 gastrointestinal perforation. Expression of VEGFR2 predicted PFS of a median of 1.4 months in VEGFR2-negative patients versus 6.4 months in VEGFR2-positive patients (P = .005). CONCLUSION Sunitinib is active in recurrent atypical/malignant meningioma patients. A randomized trial should be performed.
Collapse
Affiliation(s)
- Thomas J Kaley
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Patrick Wen
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - David Schiff
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Keith Ligon
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Sam Haidar
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Sasan Karimi
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Andrew B Lassman
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Craig P Nolan
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Igor Gavrilovic
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Andrew Norden
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Jan Drappatz
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Eudocia Quant Lee
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Benjamin Purow
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Scott R Plotkin
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Tracy Batchelor
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Lauren E Abrey
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| | - Antonio Omuro
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York (T.J.K., S.K., A.B.L., C.P.N., L.M.D., I.G., L.E.A., A.O.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachusetts (P.W., K.L., S.H., A.N., J.D., E.Q.L.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S., B.P.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (S.R.P., T.B.)
| |
Collapse
|
46
|
Wilisch-Neumann A, Pachow D, Wallesch M, Petermann A, Böhmer FD, Kirches E, Mawrin C. Re-evaluation of cytostatic therapies for meningiomas in vitro. J Cancer Res Clin Oncol 2014; 140:1343-52. [PMID: 24816784 DOI: 10.1007/s00432-014-1683-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/10/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE The purpose was to re-evaluate in cell culture models the therapeutic usefulness of some discussed chemotherapies or targeted therapies for meningiomas with a special emphasis on the role of the neurofibromatosis type 2 (NF2) tumor suppressor, which had been neglected so far. In addition, the study intended to evaluate a potential benefit from a treatment with drugs which are well established in other fields of medicine and have been linked recently with tumor disease by epidemiological studies. METHODS Meningioma cell lines corresponding to various subtypes and pairs of syngenic meningioma cell lines with or without shRNA-induced NF2 knockdown were analyzed for their dose-dependent response to the drugs in microtiter tetrazolium assays, BrdU assays and for selected cases in ELISAs measuring nucleosome liberation to specifically separate cell death from pure inhibition of cell proliferation. RESULTS We confirmed a moderate efficacy of hydroxyurea (HU) in clinically relevant concentrations. Under appropriate dosing, we neither detected major responses to the alkylating compound temozolomide nor to various drugs targeting membrane receptors or enzymes (tamoxifen, erlotinib, mifepristone, losartan, metformin and verapamil). Only concentrations far beyond achievable serum levels generated significant effects with the exception of losartan, which showed no effects at all. Chemosensitivity varied markedly among meningioma cell lines. Importantly, cells with NF2 loss exhibited a significantly higher induction of cell death by HU. CONCLUSIONS Alternative chemotherapeutic or targeted approaches besides HU have still to be evaluated in further studies, and the role of NF2 must be taken into account.
Collapse
Affiliation(s)
- Annette Wilisch-Neumann
- Department of Neuropathology, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Preusser M, Berghoff AS, Hottinger AF. High-grade meningiomas: new avenues for drug treatment? Curr Opin Neurol 2014; 26:708-15. [PMID: 24184974 DOI: 10.1097/wco.0000000000000035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW For standard first-line treatment of high-grade meningiomas, surgical resection and radiotherapy are regarded as standard of care. In the recurrent setting after exhaustion of all local treatment options, no effective therapies are known and several drugs have failed to show efficacy, but novel compounds may offer hope for better disease control. RECENT FINDINGS Upregulation of proangiogenic molecules and dysregulation of some signaling pathways such as the platelet-derived growth factor and mammalian target of rapamycin are recurrently found in high-grade meningiomas. Furthermore, in-vitro studies and single patient experience indicate that trabectedin may be an effective therapy in this tumor type. Unfortunately, so far there is a lack of conclusive clinical trials to draw definite conclusions of efficacy of these approaches. SUMMARY There remains a significant unmet need for defining the role of medical therapy in recurrent high-grade meningioma, and more basic research and multicentric well designed trials are needed in this rare and devastating tumor type. Potentially promising novel therapeutics include antiangiogenic drugs, molecular inhibitors of signaling cascades, immunotherapeutics or trabectedin. However, more basic research is required to identify more promising drug targets. VIDEO ABSTRACT AVAILABLE See the Video Supplementary Digital Content 1 (http://links.lww.com/CONR/A22).
Collapse
Affiliation(s)
- Matthias Preusser
- aDepartment of Medicine I & Comprehensive Cancer Center - CNS Unit, Medical University of Vienna bDepartment of Clinical Neurosciences, CHUV, Lausanne University Medical Center and University of Lausanne, Switzerland
| | | | | |
Collapse
|
48
|
Kaley T, Barani I, Chamberlain M, McDermott M, Panageas K, Raizer J, Rogers L, Schiff D, Vogelbaum M, Weber D, Wen P. Historical benchmarks for medical therapy trials in surgery- and radiation-refractory meningioma: a RANO review. Neuro Oncol 2014; 16:829-40. [PMID: 24500419 PMCID: PMC4022224 DOI: 10.1093/neuonc/not330] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/25/2013] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The outcomes of patients with surgery- and radiation-refractory meningiomas treated with medical therapies are poorly defined. Published reports are limited by small patient numbers, selection bias, inclusion of mixed histologic grades and stages of illness, and World Health Organization (WHO) criteria changes. This analysis seeks to define outcome benchmarks for future clinical trial design. METHODS A PubMed literature search was performed for all English language publications on medical therapy for meningioma. Reports were tabulated and analyzed for number of patients, histologic grade, prior therapy, overall survival, progression-free survival (PFS), and radiographic response. RESULTS Forty-seven publications were identified and divided by histology and prior therapies, including only those that treated patients who were surgery and radiation refractory for further analysis. This included a variety of agents (hydroxyurea, temozolomide, irinotecan, interferon-α, mifepristone, octreotide analogues, megestrol acetate, bevacizumab, imatinib, erlotinib, and gefitinib) from retrospective, pilot, and phase II studies, exploratory arms of other studies, and a single phase III study. The only outcome extractable from all studies was the PFS 6-month rate, and a weighted average was calculated separately for WHO grade I meningioma and combined WHO grade II/III meningioma. For WHO I meningioma, the weighted average PFS-6 was 29% (95% confidence interval [CI]: 20.3%-37.7%). For WHO II/III meningioma, the weighted average PFS-6 was 26% (95% CI: 19.3%-32.7%). CONCLUSIONS This comprehensive review confirms the poor outcomes of medical therapy for surgery- and radiation-refractory meningioma. We recommend the above PFS-6 benchmarks for future trial design.
Collapse
Affiliation(s)
- Thomas Kaley
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Igor Barani
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Marc Chamberlain
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Michael McDermott
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Katherine Panageas
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Jeffrey Raizer
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Leland Rogers
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - David Schiff
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Michael Vogelbaum
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Damien Weber
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| | - Patrick Wen
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York (T.K.); Department of Radiation Oncology, University of California, San Francisco, San Francisco, California (I.B.); Department of Neurology, University of Washington, Seattle, Washington (M.C.); Department of Neurosurgery, University of California, San Francisco, San Francisco, California (M.D.); Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York (K.P.); Department of Neurology, Northwestern University, Chicago, Illinois (J.R.); Department of Radiation Oncology, Gamma West Cancer Services, Salt Lake City, Utah (L.R.); Department of Neurology, University of Virginia, Charlottesville, Virginia (D.S.); Department of Neuro-Oncology, Cleveland Clinic, Cleveland, Ohio (M.V.); Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland (D.W.); Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Center, Boston, Massachsetts (P.W.)
| |
Collapse
|
49
|
Gurberg J, Bouganim N, Shenouda G, Zeitouni A. A case of recurrent anaplastic meningioma of the skull base with radiologic response to hydroxyurea. J Neurol Surg Rep 2014; 75:e52-5. [PMID: 25083390 PMCID: PMC4110141 DOI: 10.1055/s-0033-1359300] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 09/17/2013] [Indexed: 11/30/2022] Open
Abstract
Anaplastic meningiomas are rare and aggressive tumors with a high propensity for local recurrence. Surgical resection and postoperative radiotherapy are the standard of care for primary disease and local recurrences. Refractory disease is managed with chemotherapy with limited success. A highly efficacious, well-tolerated chemotherapeutic agent has yet to be found for this disease entity. Hydroxyurea is currently receiving renewed attention because of its efficacy in inducing apoptosis of meningioma cells in vitro and its favorable side-effect profile. Thus far, in humans, this agent has only induced stable disease. We describe the first patient showing a near complete/partial clinical and radiological regression after 5 months of 25 mg/kg of hydroxyurea once daily, given within 1 month after stereotactic fractionated reirradiation of a previously irradiated and operated anaplastic meningioma of the skull base. Magnetic resonance imaging showed a significant and sustained response with tumor shrinkage and cavitation.
Collapse
Affiliation(s)
- Joshua Gurberg
- Division of Otolaryngology-Head and Neck Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | | | - George Shenouda
- Department of Radiation Oncology, McGill University, Montreal, Quebec, Canada
| | - Anthony Zeitouni
- Department of Otolaryngology-Head and Neck Surgery, Royal Victoria Hospital, McGill University Health Center, Montreal, Quebec, Canada
| |
Collapse
|
50
|
A phase II trial of PTK787/ZK 222584 in recurrent or progressive radiation and surgery refractory meningiomas. J Neurooncol 2014; 117:93-101. [PMID: 24449400 DOI: 10.1007/s11060-014-1358-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 01/06/2014] [Indexed: 10/25/2022]
Abstract
When surgery and radiation are no longer treatment options, salvage systemic therapy has been used for recurrent meningiomas with little compelling evidence to suggest effectiveness. Patients with surgery and radiation refractory recurrent meningiomas were treated with the oral multifunctional tyrosine kinase inhibitor PTK787/ZK 222584 (PTK787) at a dose of 500 mg twice a day. Each treatment cycle was 4 weeks with MRI done every 8 weeks. Twenty-five patients (14 men; 11 women) with a median age of 59 years and KPS of 80 were treated. Meningioma WHO Grade was I in 2 patients, II in 14 patients and III in 8 patients; 1 patient had a hemangiopericytoma. All patients had prior surgery, external beam radiation therapy or radiosurgery and 11 patients prior systemic chemotherapy. Median number of cycles of PTK 787 administered was 4 (range <1-22). Best response in the 22 evaluable patients was stable disease in 15 (68.2 %). Predominant PTK787 related toxicities included fatigue (60 %), hypertension (24 %) and elevated transaminases (24 %). Grade II patients had a progression free survival (PFS)-6 of 64.3 %, a median PFS of 6.5 months and an overall survival (OS) of 26.0 months; grade III patients had a PFS-6 of 37.5 %, median PFS of 3.6 months and OS 23 months. PTK787 was modestly toxic at the dose of 500 mg administered twice per day. Activity as determined by PFS-6 suggests that targeting PDGF/VEGF pathway warrants further investigation.
Collapse
|