1
|
Cui J, Sun H, Sun S, Zhao H, Gu Y. Case report: Giant meningioma of the left hemisphere. Front Oncol 2024; 14:1506297. [PMID: 39711956 PMCID: PMC11659279 DOI: 10.3389/fonc.2024.1506297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024] Open
Abstract
Meningiomas are some of the most prevalent primary brain tumors in adults, and are typically non-neuroglial in nature. A variety of symptoms may be observed, including headaches, fluctuations in mental status, ataxia, muscle weakness, nausea and vomiting, seizures, visual changes, speech disorders, and sensory abnormalities. The World Health Organization (WHO) has a grading system for meningiomas based on histological criteria, which is as follows: Grade 1 meningiomas are considered benign; Grade 2 meningiomas have a moderately aggressive nature and usually present with histological atypia; and Grade 3 meningiomas exhibit aggressive malignant behavior. Grade 3 meningiomas are distinguished by aberrant and accelerated cellular proliferation, which increases the probability of invasion and recurrence within the central nervous system relative to the other grades. Malignant meningiomas are further classified by tumor size. For example, WHO grade 3 meningiomas with diameter >5 cm are designated giant meningiomas. Giant meningiomas are complicated by their potential for compression of the brain tissue, which can lead to increased intracranial pressure and hemodynamic changes. In many cases, these changes induce vasogenic edema in the adjacent brain tissue. This article details a rare case of rapidly growing atypical giant meningioma that progressed to an anterior-posterior diameter of 13 cm within 3 years, occupying the majority of the left hemisphere of the brain and encroaching upon the right intracranial structures. Through recent advances in medical diagnostics and heightened public awareness of health issues, cases with such large meningiomas have become exceedingly rare. Fortunately, the tumor in the present case was successfully resected using advanced surgical techniques that employed microscopy in conjunction with sodium fluorescein, resulting in complete removal of the tumor and restoration of the patient's muscle strength postoperatively. The value of fluorescence-guided surgery in this type of procedure is support in the present case report.
Collapse
Affiliation(s)
- Junxiang Cui
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Hu Sun
- Department of Neurosurgery, Zibo Central Hospital, Zibo, China
| | - Shuo Sun
- Department of Neurosurgery, Zibo Central Hospital, Zibo, China
| | - Hao Zhao
- Department of Neurosurgery, Zibo Central Hospital, Zibo, China
| | - Yinghao Gu
- Department of Neurosurgery, Zibo Central Hospital, Zibo, China
| |
Collapse
|
2
|
Cossu G, Le Van T, Kerherve L, Houidi SA, Morlaix E, Bonneville F, Chapon R, Baland O, Cao C, Lleu M, Farah W, El Cadhi A, Beaurain J, Picart T, Xu B, Berhouma M. Enlightening the invisible: Applications, limits and perspectives of intraoperative fluorescence in neurosurgery. BRAIN & SPINE 2024; 4:103928. [PMID: 39823065 PMCID: PMC11735926 DOI: 10.1016/j.bas.2024.103928] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 01/19/2025]
Abstract
Introduction The introduction of intraoperative fluorophores represented a significant advancement in neurosurgical practice. Nowadays they found different applications: in oncology to improve the visualization of tumoral tissue and optimize resection rates and in vascular neurosurgery to assess the exclusion of vascular malformations or the permeability of bypasses, with real-time intraoperative evaluations. Research question A comprehensive knowledge of how fluorophores work is crucial to maximize their benefits and to incorporate them into daily neurosurgical practice. We would like to revise here their applications and clinical relevance. Material and methods A focused literature review of relevant articles dealing with the versatile applications of fluorophores in neurosurgery was performed. Results The fundamental mechanisms of action of intraoperative fluorophores are enlightened, examining their interactions with target tissues and the principles driving fluorescence-guided surgery. The clinical applications of the principal fluorophores, namely fluorescein sodium, 5-ALA and indocyanine green, are detailed, in regards to the management of vascular malformations, brain tumors and pathologies treated through endoscopic endonasal approaches. Discussion and conclusion Future perspective dealing with the development of new technologies or of new molecules are discussed. By critically assessing the efficacy and applications of the different fluorophores, as well as charting their potential future uses, this paper seeks to guide clinicians in their practice and provide insights for driving innovation and progress in fluorescence-based surgery and research.
Collapse
Affiliation(s)
- Giulia Cossu
- Department of Neurosurgery, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Tuan Le Van
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Luc Kerherve
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Sayda A. Houidi
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Edouard Morlaix
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Florent Bonneville
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Renan Chapon
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Olivier Baland
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Catherine Cao
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Maxime Lleu
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Walid Farah
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Ahmed El Cadhi
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Jacques Beaurain
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
| | - Thiebaud Picart
- Department of Neurosurgery, Groupe Hospitalier Est, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
- Université Claude Bernard Lyon 1, 43 Bd du 11 Novembre 1918, Villeurbanne, France
- Cancer Research Centre of Lyon (CRCL), INSERM 1052, CNRS 5286, 28 Rue Laennec, Lyon, France
| | - Bin Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Moncef Berhouma
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France
- Functional and Molecular Imaging Team (CNRS 6302), Molecular Chemistry Institute (ICMUB), University of Burgundy, France
| |
Collapse
|
3
|
de Laurentis C, Pasquali C, Beuriat PA, Mottolese C, Szathmari A, Vinchon M, Di Rocco F. Role of sodium fluorescein in pediatric low-grade glioma surgery: an update. Childs Nerv Syst 2024; 40:3143-3154. [PMID: 38662222 DOI: 10.1007/s00381-024-06402-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE Complete surgical resection is still the mainstay in the treatment of central nervous system low-grade tumors, eventually resulting curative. The complete surgical removal of these lesions, however, may be difficult in some cases because of their infiltrative nature. Intraoperative adjuncts may be a game changer. Sodium fluorescein (SF) is among the ideal candidates as intraoperative tools to favor the actual recognition of the tumor extension, since it accumulates in areas of altered blood-brain barrier, a typical characteristic of pediatric gliomas, and has a low rate of adverse events. This work proposes an update of previous works about the evaluation of the feasibility and usefulness of a systematic use of SF in a low-grade lesion group of pediatric patients. METHODS Pediatric patients operated on for a resection or a biopsy of a low-grade glial or glioneuronal lesion (WHO grade I and II) at our Institution between September 2021 and December 2023, with the intraoperative use of sodium fluorescein (SF), were enrolled in the study. We collected pre-operative and postoperative clinical and radiological data, intraoperative findings, and post-operative pathological diagnoses. RESULTS No adverse events were registered related to the intraoperative use of SF. SF appeared useful for the localization of boundaries of tumors, especially when characterized by a high degree of infiltration or by a deep-seated location, and for the checking of possible tumor remnants at the end of surgery. A good tumor-to-healthy tissue contrast was registered when tumor visualization was in a range between 1 to 2 h and 30 min after SF injection. Possible "false positives" due to intraoperative vascular wall injury and clearance of SF from both tumor and healthy tissue were observed in some cases and still remain open issues. CONCLUSIONS SF is a feasible and safe intraoperative adjunct tool in the surgical removal of pediatric low-grade tumors. SF may show its usefulness especially in selected cases, such as deep-seated lesions and infiltrating tumors. Its safety profile, user-friendly management, and potential utility in both tumor resections and neuronavigated biopsies favor its wider use in the surgical treatment of pediatric low-grade tumors.
Collapse
Affiliation(s)
- Camilla de Laurentis
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
- Functional Neurosurgery Unit, Hôpital Pierre Wertheimer, Lyon, France
| | - Claudia Pasquali
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
| | - Pierre-Aurélien Beuriat
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
- Functional Neurosurgery Unit, Hôpital Pierre Wertheimer, Lyon, France
- Department of Medicine and Surgery, Université Claude Bernard, Lyon 1, Lyon, France
| | - Carmine Mottolese
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
| | - Alexandru Szathmari
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
- Functional Neurosurgery Unit, Hôpital Pierre Wertheimer, Lyon, France
| | - Matthieu Vinchon
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
| | - Federico Di Rocco
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France.
- Department of Medicine and Surgery, Université Claude Bernard, Lyon 1, Lyon, France.
| |
Collapse
|
4
|
Kırış T, Akçakaya MO. Comparison of intraoperative sodium fluorescein and indocyanine green videoangiography during intracranial aneurysm and arteriovenous malformation surgery. Clin Neurol Neurosurg 2024; 244:108414. [PMID: 39002271 DOI: 10.1016/j.clineuro.2024.108414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/13/2024] [Accepted: 06/28/2024] [Indexed: 07/15/2024]
Abstract
OBJECTIVE Indocyanine green (ICG) and sodium fluorescein (Na-Fl) are two fluorophores, which are used for videoangiography purposes. This prospective study reports our experience by using surgical microscopes equipped with two special filters. We compared the imaging efficacy of Na-FL and ICG videoangiography techniques during aneurysm and arteriovenous malformations (AVM) surgeries. PATIENT AND METHODS Fourtynine consecutive patients were operated between September 2015 and December 2022. Patients with ruptured/unruptured aneurysms or with AVMs presented with/without hemorrhage were included to the current study. RESULTS There were a total of 48 aneurysms and 11 AVMs in 49 patients in the current study. Na-Fl enables the surgeon to manipulate vessels and aneurysms real-time. ICG provides the ability to watch the videoangiography repeatedly and understand the angioarchitecture. The use of FLOW-800 module allows the surgeon to make concrete judgements about the flow dynamics of AVMs and therefore ICG videoangiography found useful in AVM surgery. Na-Fl has been found more useful for the evalution of the low caliber perforating arteries and anatomical features of the aneursym dome in aneurysm surgery. CONCLUSION The results of our series demonstrated the safety and efficacy of Na-Fl use in the surgery of aneurysms and AVMs. Both the ICG and Na-Fl videoangiographies are easy to perform and complimentary to each other. Combined use of these two mediums may provide better surgical results, considering the separate advantages and disadvantages of these both techniques.
Collapse
Affiliation(s)
- Talat Kırış
- Department of Neurosurgery, American Hospital, Istanbul, Turkey
| | - Mehmet Osman Akçakaya
- Department of Neurosurgery, Gayrettepe Florence Nightingale Hospital, Istanbul, Turkey.
| |
Collapse
|
5
|
Suero Molina E, Bruneau M, Reuter G, Shahein M, Cavallo LM, Daniel RT, Kasper EM, Froelich S, Jouanneau E, Manet R, Messerer M, Mazzatenta D, Meling TR, Roche PH, Schroeder HWS, Tatagiba M, Visocchi M, Prevedello DM, Stummer W, Cornelius JF, EANS Skull Base Section. Fluorescence guidance in skull base surgery: Applications and limitations - A systematic review. BRAIN & SPINE 2024; 4:103328. [PMID: 39309550 PMCID: PMC11416557 DOI: 10.1016/j.bas.2024.103328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024]
Abstract
Introduction Intraoperative fluorescence guidance is a well-established surgical adjunct in high-grade glioma surgery. In contrast, the clinical use of such dyes and technology has been scarcely reported in skull base surgery. Research question We aimed to systematically review the clinical applications of different fluorophores in both open and endonasal skull base surgery. Material and methods We performed a systematic review and discussed the current literature on fluorescence guidance in skull base surgery. Results After a comprehensive literature search, 77 articles on skull base fluorescence guidance were evaluated. A qualitative analysis of the articles is presented, discussing clinical indications and current controversies. The use of intrathecal fluorescein was the most frequently reported in the literature. Beyond that, 5-ALA and ICG were two other fluorescent dyes most extensively discussed, with some experimental fluorophore applications in skull base surgery. Discussion and conclusion Intraoperative fluorescence imaging can serve as an adjunct technology in skull base surgery. The scope of initial indications of these fluorophores has expanded beyond malignant glioma resection alone. We discuss current use and controversies and present an extensive overview of additional indications for fluorescence imaging in skull base pathologies. Further quantitative studies will be needed in the future, focusing on tissue selectivity and time-dependency of the different fluorophores currently commercially available, as well as the development of new compounds to expand applications and facilitate skull base surgeries.
Collapse
Affiliation(s)
- Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Michael Bruneau
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Gilles Reuter
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | | | - Luigi M. Cavallo
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, Federico II University of Naples, Policlinico Federico II University Hospital, Italy
| | - Roy T. Daniel
- Department of Neurosurgery, Department of Neuroscience, Centre Hospitalier Universitaire Vaudois, University Hospital Lausanne, Switzerland
| | - Ekkehard M. Kasper
- Department of Neurosurgery, Boston University Medical School, MA and Steward Medical Group, Brighton, MA/USA McMaster University Faculty of Health Sciences, Hamilton, ON, Canada
| | - Sebastien Froelich
- Department of Neurosurgery, Lariboisière Hospital, Université Paris Diderot, Paris, France
| | - Emanuel Jouanneau
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Lyon, France
| | - Romain Manet
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Lyon, France
| | - Mahmoud Messerer
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, Federico II University of Naples, Policlinico Federico II University Hospital, Italy
| | - Diego Mazzatenta
- Department of Neurosurgery, Neurological Sciences Institut IRCCS, Bologna, Italy
| | - Torstein R. Meling
- Department of Neurosurgery, The National Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pierre-Hugues Roche
- Department of Neurosurgery, Aix-Marseille Université, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Marseille, France
| | | | - Marcos Tatagiba
- Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | - Massimiliano Visocchi
- Department of Neurosurgery, Institute of Neurosurgery Catholic University of Rome, Italy
| | - Daniel M. Prevedello
- Deparmtent of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| | - Jan F. Cornelius
- Department of Neurosurgery, University Hospital of Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - EANS Skull Base Section
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
- Department of Neurosurgery, Mansoura University, Egypt
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, Federico II University of Naples, Policlinico Federico II University Hospital, Italy
- Department of Neurosurgery, Department of Neuroscience, Centre Hospitalier Universitaire Vaudois, University Hospital Lausanne, Switzerland
- Department of Neurosurgery, Boston University Medical School, MA and Steward Medical Group, Brighton, MA/USA McMaster University Faculty of Health Sciences, Hamilton, ON, Canada
- Department of Neurosurgery, Lariboisière Hospital, Université Paris Diderot, Paris, France
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Lyon, France
- Department of Neurosurgery, Neurological Sciences Institut IRCCS, Bologna, Italy
- Department of Neurosurgery, The National Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Neurosurgery, Aix-Marseille Université, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Marseille, France
- Department of Neurosurgery, University Medicine Greifswald, Germany
- Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany
- Department of Neurosurgery, Institute of Neurosurgery Catholic University of Rome, Italy
- Deparmtent of Neurosurgery, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Neurosurgery, University Hospital of Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
6
|
Hsieh AL, Bi WL, Ramesh V, Brastianos PK, Plotkin SR. Evolving concepts in meningioma management in the era of genomics. Cancer 2024; 130:2586-2600. [PMID: 38753473 PMCID: PMC11260245 DOI: 10.1002/cncr.35279] [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: 04/08/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 05/18/2024]
Abstract
Meningioma is the most common type of primary brain tumor. Surgical resection followed by surveillance is the first-line treatment for the majority of symptomatic meningiomas; however, recent advances in molecular sequencing, DNA methylation, proteomics, and single-cell sequencing provide insights into further characterizing this heterogeneous group of tumors with a wide range of prognoses. A subset of these tumors are highly aggressive and cause severe morbidity and mortality. Therefore, identifying those individuals with a poor prognosis and intervening are critical. This review aims to help readers interpret the molecular profiling of meningiomas to identify patients with worse prognoses and guide the management and strategy for surveillance.
Collapse
Affiliation(s)
- Annie L. Hsieh
- Department of Neurology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Vijaya Ramesh
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Priscilla K Brastianos
- Department of Neurology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Central Nervous System Metastasis Program, Massachusetts General Hospital, Boston, MA, United States
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Scott R. Plotkin
- Department of Neurology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Wagner A, Brielmaier MC, Kampf C, Baumgart L, Aftahy AK, Meyer HS, Kehl V, Höhne J, Schebesch KM, Schmidt NO, Zoubaa S, Riemenschneider MJ, Ratliff M, Enders F, von Deimling A, Liesche-Starnecker F, Delbridge C, Schlegel J, Meyer B, Gempt J. Fluorescein-stained confocal laser endomicroscopy versus conventional frozen section for intraoperative histopathological assessment of intracranial tumors. Neuro Oncol 2024; 26:922-932. [PMID: 38243410 PMCID: PMC11066924 DOI: 10.1093/neuonc/noae006] [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: 10/10/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND The aim of this clinical trial was to compare Fluorescein-stained intraoperative confocal laser endomicroscopy (CLE) of intracranial lesions and evaluation by a neuropathologist with routine intraoperative frozen section (FS) assessment by neuropathology. METHODS In this phase II noninferiority, prospective, multicenter, nonrandomized, off-label clinical trial (EudraCT: 2019-004512-58), patients above the age of 18 years with any intracranial lesion scheduled for elective resection were included. The diagnostic accuracies of both CLE and FS referenced with the final histopathological diagnosis were statistically compared in a noninferiority analysis, representing the primary endpoint. Secondary endpoints included the safety of the technique and time expedited for CLE and FS. RESULTS A total of 210 patients were included by 3 participating sites between November 2020 and June 2022. Most common entities were high-grade gliomas (37.9%), metastases (24.1%), and meningiomas (22.7%). A total of 6 serious adverse events in 4 (2%) patients were recorded. For the primary endpoint, the diagnostic accuracy for CLE was inferior with 0.87 versus 0.91 for FS, resulting in a difference of 0.04 (95% confidence interval -0.10; 0.02; P = .367). The median time expedited until intraoperative diagnosis was 3 minutes for CLE and 27 minutes for FS, with a mean difference of 27.5 minutes (standard deviation 14.5; P < .001). CONCLUSIONS CLE allowed for a safe and time-effective intraoperative histological diagnosis with a diagnostic accuracy of 87% across all intracranial entities included. The technique achieved histological assessments in real time with a 10-fold reduction of processing time compared to FS, which may invariably impact surgical strategy on the fly.
Collapse
Affiliation(s)
- Arthur Wagner
- Department of Neurosurgery, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Maria Charlotte Brielmaier
- Department of Neurosurgery, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Charlotte Kampf
- Department of Neurosurgery, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Lea Baumgart
- Department of Neurosurgery, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Amir Kaywan Aftahy
- Department of Neurosurgery, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Hanno S Meyer
- Department of Neurosurgery, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Victoria Kehl
- Institute for AI and Informatics in Medicine & Muenchner Studienzentrum (MSZ), Technical University Munich School of Medicine, Munich, Germany
| | - Julius Höhne
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany
- Department of Neurosurgery, Paracelsus Medical University, Nürnberg, Germany
| | - Karl-Michael Schebesch
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany
- Department of Neurosurgery, Paracelsus Medical University, Nürnberg, Germany
| | - Nils O Schmidt
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany
| | - Saida Zoubaa
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | | | - Miriam Ratliff
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany
| | - Frederik Enders
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany
| | - Andreas von Deimling
- Department of Neuropathology, University Hospital Heidelberg and CCU Neuropathology, German Cancer Center (DKFZ), Heidelberg, Germany
| | | | - Claire Delbridge
- Department of Neuropathology, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Juergen Schlegel
- Department of Neuropathology, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar Technical University Munich School of Medicine, Munich, Germany
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
8
|
Diehl CD, Giordano FA, Grosu AL, Ille S, Kahl KH, Onken J, Rieken S, Sarria GR, Shiban E, Wagner A, Beck J, Brehmer S, Ganslandt O, Hamed M, Meyer B, Münter M, Raabe A, Rohde V, Schaller K, Schilling D, Schneider M, Sperk E, Thomé C, Vajkoczy P, Vatter H, Combs SE. Opportunities and Alternatives of Modern Radiation Oncology and Surgery for the Management of Resectable Brain Metastases. Cancers (Basel) 2023; 15:3670. [PMID: 37509330 PMCID: PMC10377800 DOI: 10.3390/cancers15143670] [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: 04/18/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Postsurgical radiotherapy (RT) has been early proven to prevent local tumor recurrence, initially performed with whole brain RT (WBRT). Subsequent to disadvantageous cognitive sequalae for the patient and the broad distribution of modern linear accelerators, focal irradiation of the tumor has omitted WBRT in most cases. In many studies, the effectiveness of local RT of the resection cavity, either as single-fraction stereotactic radiosurgery (SRS) or hypo-fractionated stereotactic RT (hFSRT), has been demonstrated to be effective and safe. However, whereas prospective high-level incidence is still lacking on which dose and fractionation scheme is the best choice for the patient, further ablative techniques have come into play. Neoadjuvant SRS (N-SRS) prior to resection combines straightforward target delineation with an accelerated post-surgical phase, allowing an earlier start of systemic treatment or rehabilitation as indicated. In addition, low-energy intraoperative RT (IORT) on the surgical bed has been introduced as another alternative to external beam RT, offering sterilization of the cavity surface with steep dose gradients towards the healthy brain. This consensus paper summarizes current local treatment strategies for resectable brain metastases regarding available data and patient-centered decision-making.
Collapse
Affiliation(s)
- Christian D Diehl
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 München, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 80336 München, Germany
| | - Frank A Giordano
- Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, University Medical Center, Medical Faculty, 79106 Freiburg, Germany
| | - Sebastian Ille
- Department of Neurosurgery, Faculty of Medicine, Technical University of Munich, 81675 München, Germany
| | - Klaus-Henning Kahl
- Department of Radiation Oncology, University Medical Center Augsburg, 86156 Augsburg, Germany
| | - Julia Onken
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiotherapy and Radiation Oncology, University Medical Center Göttingen, 37075 Göttingen, Germany
- Comprehensive Cancer Center Niedersachsen (CCC-N), 37075 Göttingen, Germany
| | - Gustavo R Sarria
- Department of Radiation Oncology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Ehab Shiban
- Department of Neurosurgery, University Medical Center Augsburg, 86156 Augsburg, Germany
| | - Arthur Wagner
- Department of Neurosurgery, Faculty of Medicine, Technical University of Munich, 81675 München, Germany
| | - Jürgen Beck
- Department of Neurosurgery, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Stefanie Brehmer
- Department of Neurosurgery, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Oliver Ganslandt
- Neurosurgical Clinic, Klinikum Stuttgart, 70174 Stuttgart, Germany
| | - Motaz Hamed
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Faculty of Medicine, Technical University of Munich, 81675 München, Germany
| | - Marc Münter
- Department of Radiation Oncology, Klinikum Stuttgart Katharinenhospital, 70174 Stuttgart, Germany
| | - Andreas Raabe
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Veit Rohde
- Department of Neurosurgery, Universitätsmedizin Göttingen, 37075 Göttingen, Germany
| | - Karl Schaller
- Department of Neurosurgery, University of Geneva Medical Center & Faculty of Medicine, 1211 Geneva, Switzerland
| | - Daniela Schilling
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 München, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Matthias Schneider
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Elena Sperk
- Mannheim Cancer Center, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 München, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 80336 München, Germany
| |
Collapse
|
9
|
de Laurentis C, Bteich F, Beuriat PA, Mottolese C, Giussani C, Szathmari A, Vinchon M, Di Rocco F. Sodium fluorescein in pediatric neurosurgery: a systematic review with technical considerations and future perspectives. Childs Nerv Syst 2022; 39:1451-1462. [PMID: 36459209 DOI: 10.1007/s00381-022-05772-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Sodium fluorescein (SF) is routinely used in several centers as a valid intraoperative adjunct in adult oncological neurosurgery. Its use in pediatric neurosurgery is increasing, although its role is not yet well-defined in children. We reviewed the current literature in order to evaluate the use of SF in children with CNS and PNS lesions. METHODS For this systematic review, we searched PubMed, Scopus, and Embase databases, and forward and backward citations for studies published between database inception and July 31st, 2022. We included any article type or congress abstract adding at least a new case, without restrictions of language or publication status, concerning the use of SF in neurosurgical procedures in patients under 18 years of age. We excluded studies concerning purely vascular cases and cerebrospinal fluid leaks. RESULTS Of 4094 records identified, 19 articles were eligible and included for further analysis. As per July 31st, 2022, at least 119 patients aged from 11 months to 17.9 years underwent surgery with SF. No serious adverse events were reported. A large variety of tumor types was operated, in most cases resected under the specific YELLOW 560 nm filter after a low-dose SF injection (2-5 mg/kg) at the end of anesthesia induction. SF was reported particularly useful in gangliogliomas and pilocytic astrocytomas. DISCUSSION/CONCLUSION Given its easy-to-use profile, low cost, and safety, SF seems to be a feasible and valid adjunct in the pediatric population when aiming at individuating a biopsy target or maximizing extent of resection, particularly in some tumor types. Further studies are required to strengthen the evidence on its impact on outcomes.
Collapse
Affiliation(s)
- Camilla de Laurentis
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France. .,Department of Medicine and Surgery, Università degli Studi di Milano Bicocca, Milan, Italy.
| | - Fred Bteich
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
| | | | - Carmine Mottolese
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
| | - Carlo Giussani
- Department of Medicine and Surgery, Università degli Studi di Milano Bicocca, Milan, Italy.,Neurosurgery Unit, San Gerardo Hospital, Monza, Italy
| | | | - Matthieu Vinchon
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France
| | - Federico Di Rocco
- Pediatric Neurosurgery Unit, Hôpital Femme Mère Enfant, Lyon, France.,Department of Medicine and Surgery, Université Claude Bernard Lyon I, Lyon, France
| |
Collapse
|
10
|
Muto J, Mine Y, Nishiyama Y, Murayama K, Yamada S, Kojima D, Hayakawa M, Adachi K, Hasegawa M, Lee JYK, Hirose Y. Intraoperative Real-Time Near-Infrared Image-Guided Surgery to Identify Intracranial Meningiomas via Microscope. Front Neurosci 2022; 16:837349. [PMID: 35600609 PMCID: PMC9114498 DOI: 10.3389/fnins.2022.837349] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/24/2022] Open
Abstract
Meningiomas are a common pathology in the central nervous system requiring complete surgical resection. However, in cases of recurrence and post-irradiation, accurate identification of tumor remnants and a dural tail under bright light remains challenging. We aimed to perform real-time intraoperative visualization of the meningioma and dural tail using a delayed-window indocyanine green (ICG) technique with microscopy. Fifteen patients with intracranial meningioma received 0.5 mg/kg ICG a few hours before observation during the surgery. We used near-infrared (NIR) fluorescence to identify the tumor location. NIR fluorescence could visualize meningiomas in 12 out of 15 cases. Near-infrared visualization during the surgery ranged from 1 to 4 h after the administration of ICG. The mean signal-to-background ratio (SBR) of the intracranial meningioma in delayed-window ICG (DWIG) was 3.3 ± 2.6. The ratio of gadolinium-enhanced T1 tumor signal to the brain (T1BR) (2.5 ± 0.9) was significantly correlated with the tumor SBR (p = 0.016). Ktrans, indicating blood–brain barrier permeability, was significantly correlated with tumor SBR (p < 0.0001) and T1BR (p = 0.013) on dynamic contrast-enhanced magnetic resonance imaging (MRI). DWIG demonstrated a sensitivity of 94%, specificity of 38%, positive predictive value (PPV) of 76%, and negative predictive value (NPV) of 75% for meningiomas. This is the first pilot study in which DWIG fluorescence-guided surgery was used to visualize meningioma and dural tail intraoperatively with microscopy. DWIG is comparable with second-window ICG in terms of mean SBR. Gadolinium-enhanced T1 tumor signal may predict NIR fluorescence of the intracranial meningioma. Blood–brain barrier permeability as shown by Ktrans on dynamic contrast-enhanced MRI can contribute to gadolinium enhancement on MRI and to ICG retention and tumor fluorescence by NIR.
Collapse
Affiliation(s)
- Jun Muto
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
- *Correspondence: Jun Muto,
| | - Yutaka Mine
- Department of Neurosurgery, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan
| | - Yuya Nishiyama
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | | | - Seiji Yamada
- Department of Pathology, Fujita Health University, Toyoake, Japan
| | - Daijiro Kojima
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | - Motoharu Hayakawa
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | - Kazuhide Adachi
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | | | - John Y. K. Lee
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| |
Collapse
|
11
|
Schwartz TH, McDermott MW. The Simpson grade: abandon the scale but preserve the message. J Neurosurg 2021; 135:488-495. [PMID: 33035995 DOI: 10.3171/2020.6.jns201904] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/16/2020] [Indexed: 11/06/2022]
Abstract
The Simpson grading scale, developed in 1957 by Donald Simpson, has been considered the gold standard for defining the surgical extent of resection for WHO grade I meningiomas. Since its introduction, the scale and its modifications have generated enormous controversy. The Simpson grade is based on an intraoperative visual assessment of resection, which is subjective and notoriously inaccurate. The majority of studies in which the grading system was used were performed before routine postoperative MRI surveillance was employed, rendering assessments of extent of resection and the definition of recurrence inconsistent. The infiltration and proliferation potential of tumor components such as hyperostotic bone and dural tail vary widely based on tumor location, as does the molecular biology of the tumor, rendering a universal scale for all meningiomas unfeasible. While extent of resection is clearly important at reducing recurrence rates, achieving the highest Simpson grade resection should not always be the goal of surgery. Donald Simpson's name and his scale deserve to be recognized and preserved in the historical pantheon of pioneering and transformative neurosurgical concepts. Nevertheless, his eponymous scale is no longer relevant in modern meningioma surgery. While his message of maximizing extent of resection and minimizing morbidity is still germane, a single measure using subjective criteria cannot be applied universally to all meningiomas, regardless of location. Meningioma surgery should be performed with the goal of achieving maximal safe resection, ideally guided by molecularly tagged fluorescent labeling and assessed using objective criteria, including postoperative MRI as well as molecularly tagged scans such as [68Ga]-DOTATATE-PET.
Collapse
Affiliation(s)
- Theodore H Schwartz
- 1Department of Neurosurgery, Otolaryngology and Neuroscience, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, New York; and
| | - Michael W McDermott
- 2Division of Neuroscience, Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
12
|
Schupper AJ, Rao M, Mohammadi N, Baron R, Lee JYK, Acerbi F, Hadjipanayis CG. Fluorescence-Guided Surgery: A Review on Timing and Use in Brain Tumor Surgery. Front Neurol 2021; 12:682151. [PMID: 34220688 PMCID: PMC8245059 DOI: 10.3389/fneur.2021.682151] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
Fluorescence-guided surgery (FGS) allows surgeons to have improved visualization of tumor tissue in the operating room, enabling maximal safe resection of malignant brain tumors. Over the past two decades, multiple fluorescent agents have been studied for FGS, including 5-aminolevulinic acid (5-ALA), fluorescein sodium, and indocyanine green (ICG). Both non-targeted and targeted fluorescent agents are currently being used in clinical practice, as well as under investigation, for glioma visualization and resection. While the efficacy of intraoperative fluorescence in studied fluorophores has been well established in the literature, the effect of timing on fluorophore administration in glioma surgery has not been as well depicted. In the past year, recent studies of 5-ALA use have shown that intraoperative fluorescence may persist beyond the previously studied window used in prior multicenter trials. Additionally, the use of fluorophores for different brain tumor types is discussed in detail, including a discussion of choosing the right fluorophore based on tumor etiology. In the following review, the authors will describe the temporal nature of the various fluorophores used in glioma surgery, what remains uncertain in FGS, and provide a guide for using fluorescence as a surgical adjunct in brain tumor surgery.
Collapse
Affiliation(s)
- Alexander J Schupper
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Manasa Rao
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nicki Mohammadi
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rebecca Baron
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - John Y K Lee
- Department of Neurosurgery, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Francesco Acerbi
- Department of Neurosurgery, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | | |
Collapse
|
13
|
Fluorescence image-guided resection of intracranial meningioma: an experimental in vivo study on nude mice. Ann Anat 2021; 237:151752. [PMID: 33940118 DOI: 10.1016/j.aanat.2021.151752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The use of photodynamic agents in malignant cranial tumor surgery is quite common. For example five-aminolevulinic acid (5-ALA)-induced porphyrins in malignant gliomas are potent photosensitizers. Until today there is no comparable selective fluorescent substance available for meningiomas. Nevertheless, there is a demand for intraoperative fluorescent identification of e.g. invasive skull base meningiomas to increase radicality. This study was established to investigate fluorescent image-guided resection with somatostatin receptor labelled fluorescence dye for intracranial meningioma in the nude mice. METHODS Primary meningioma cell culture samples were stereotactically implanted subdural into 20 nude mice. 90 days after inoculation of the cells, a cranial MRI with contrast agent revealed tumor growth. After detection of tumor mass in MRI, FAM-TOC5,6-Carboxyfluoresceine-Tyr3-Octreotide was injected intravenously and tumor mass was hereafter resected under visualization via fluorescence microscope and endoscope. After attempted total resection, animal were sacrificed brain slices were obtained and histologically analysed to verify the resection extent. RESULTS In 18 mice tumor growth was detected in MRI after 90 days of inoculation. The tumor mass could be clearly identified with fluorescence microscope and endoscope after injecting FAM-TOC5,6-Carboxyfluoresceine-Tyr3-Octreotide. The tumor margins could be better visualized. After fluorescence-guided resection no remaining tumor could be identified in histological analysis. CONCLUSIONS This study describes for the first time the use of FAM-TOC5,6-Carboxyfluoresceine-Tyr3-Octreotide and demonstrates its value of fluorescent identification of meningioma cells in vivo. Furthermore, the authors established a new experimental animal model for fluorescence meningioma surgery.
Collapse
|
14
|
Olguner SK, Arslan A, Açık V, İstemen İ, Can M, Gezercan Y, Ökten Aİ. Sodium Fluorescein for Spinal Intradural Tumors. Front Oncol 2021; 10:618579. [PMID: 33585245 PMCID: PMC7877540 DOI: 10.3389/fonc.2020.618579] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/11/2020] [Indexed: 01/11/2023] Open
Abstract
Technological innovations in spinal intradural tumor surgery simplify treatment. Surgical treatment of cranial benign and malignant pathologies under microscope with sodium (Na)-fluorescein guidance has often been reported, but few studies have focused on spinal intradural tumors. We aimed to investigate the usefulness of Na-fluorescein under yellow filter in intradural spinal tumor surgery by retrospectively reviewing cases involving intramedullary and extramedullary tumors operated under the guidance of Na-fluorescein. Forty-nine adult patients with a diagnosis of spinal intradural tumor operated under a yellow filter (560 nm) microscope using Na-fluorescein dye were included in the study. Demographic data, such as age and sex, neurological status, extent of tumor resection, histopathological diagnosis, Na-fluorescein staining pattern, and its usefulness during surgery were noted and statistically analyzed. Of all recruited patients, 26 women (53.1%) and 23 men (46.9%), were included for analysis. The age range of the patients was 18–64 years, with a mean age of 41.6 ± 13.9. An intradural intramedullary mass was found in 30.6% (n = 15) of the patients, and an intradural extramedullary mass in 69.4% (n: 34). While Na-fluorescein staining was homogeneous in all intradural extramedullary tumors, 73.3% (n: 11) of intradural intramedullary tumors were homogeneous, and 13.3% (n: 2) moderately heterogeneous. In the whole study group, the Na-fluorescein staining pattern was helpful in surgical resection in 47 cases (95.9%). While 34/34 (100%) found it helpful for extramedullary tumors, 13/15 (86.7%) did in intramedullary tumors, and for 2/15 (13.3%) it was not. In conclusion, Na-fluorescein helps in distinguishing tumor from healthy tissue in intradural extramedullary and intramedullary tumor surgery under a yellow filter microscope in most cases, thus providing convenient assistance to surgeons.
Collapse
Affiliation(s)
- Semih Kivanc Olguner
- Department of Neurosurgery, Adana City Training and Research Hospital, Adana, Turkey
| | - Ali Arslan
- Department of Neurosurgery, Adana City Training and Research Hospital, Adana, Turkey
| | - Vedat Açık
- Department of Neurosurgery, Adana City Training and Research Hospital, Adana, Turkey
| | - İsmail İstemen
- Department of Neurosurgery, Adana City Training and Research Hospital, Adana, Turkey
| | - Mehmet Can
- Department of Neurosurgery, Adana City Training and Research Hospital, Adana, Turkey
| | - Yurdal Gezercan
- Department of Neurosurgery, Adana City Training and Research Hospital, Adana, Turkey
| | - Ali İhsan Ökten
- Department of Neurosurgery, Adana City Training and Research Hospital, Adana, Turkey
| |
Collapse
|
15
|
Höhne J, Schebesch KM, Zoubaa S, Proescholdt M, Riemenschneider MJ, Schmidt NO. Intraoperative imaging of brain tumors with fluorescein: confocal laser endomicroscopy in neurosurgery. Clinical and user experience. Neurosurg Focus 2021; 50:E19. [PMID: 33386020 DOI: 10.3171/2020.11.focus20783] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/02/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Confocal laser endomicroscopy (CLE) is an established tool in basic research for tissue imaging at the level of microstructures. Miniaturization and refinement of the technology have made this modality available for operative imaging with a handheld device. Sufficient image contrast is provided by the preoperative application of fluorescein sodium. The authors report their first experiences in a clinical case series using the new confocal laser endomicroscope. METHODS Handling, operative workflow, and visualization of the CLE were critically evaluated in 12 cases of different CNS tumors. Three different imaging positions in relation to the tumor were chosen: the tumor border (I), tumor center (II), and perilesional zone (III). Respective diagnostic sampling with H & E staining and matching intraoperative neuronavigation and microscope images are provided. RESULTS CLE was found to be beneficial in terms of high-quality visualization of fine structures and for displaying hidden anatomical details. The handling of the device was good, and the workflow was easy. CONCLUSIONS Handling ergonomics and image acquisition are intuitive. The endomicroscope allows excellent additional visualization of microstructures in the surgical field with a minimally invasive technique and could improve safety and clinical outcomes. The new confocal laser endomicroscope is an advanced tool with the potential to change intracranial tumor surgery. Imaging of these microstructures is novel, and research with comparative validation with traditional neuropathological assessments is needed.
Collapse
Affiliation(s)
- Julius Höhne
- 1Department of Neurosurgery.,3Wilhelm Sander-Neuro-Oncology Unit, University Medical Center Regensburg, Germany
| | - Karl-Michael Schebesch
- 1Department of Neurosurgery.,3Wilhelm Sander-Neuro-Oncology Unit, University Medical Center Regensburg, Germany
| | - Saida Zoubaa
- 2Institute of Neuropathology, and.,3Wilhelm Sander-Neuro-Oncology Unit, University Medical Center Regensburg, Germany
| | - Martin Proescholdt
- 1Department of Neurosurgery.,3Wilhelm Sander-Neuro-Oncology Unit, University Medical Center Regensburg, Germany
| | - Markus J Riemenschneider
- 2Institute of Neuropathology, and.,3Wilhelm Sander-Neuro-Oncology Unit, University Medical Center Regensburg, Germany
| | - Nils Ole Schmidt
- 1Department of Neurosurgery.,3Wilhelm Sander-Neuro-Oncology Unit, University Medical Center Regensburg, Germany
| |
Collapse
|
16
|
Sánchez Fernández C, Choque Cuba B, Rivero-Garvía M, de Borja Arteaga Romero F, Márquez Rivas J. Combined microsurgical fluorescence for optimizing resection in refractory empyema and cerebritis. Childs Nerv Syst 2020; 36:1835-1841. [PMID: 32601901 DOI: 10.1007/s00381-020-04762-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/22/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE Due to the generalization of new microsurgical equipment, intraoperative fluorescence techniques have extended in neurosurgical practice, mainly in neurovascular and neuro-oncology patients. The aim of identifying pathological tissue and also differentiating from the normal brain helps neurosurgeons to approach other kinds of intracranial entities such as infections. METHODS It is described in the case of an 11-year-old patient who underwent a subdural empyema by performing a craniotomy and evacuation of the purulent collection. After a non-optimal evolution, a frontobasal meningoencephalitis was assessed with cerebral involvement and associated intracranial hypertension. Indocyanine green (ICG) was used in reintervention for demonstrating a great damage of cortical vascularization around the infected area as well as fluorescein (FL), which identified a large area of avascularized tissue. RESULTS Both techniques allowed a selective excision of the affected brain parenchyma while preserving viable parenchymal areas. Radiological evolution and clinical outcome were good. CONCLUSIONS The identification of vascular patterns in brain lesions and the recognition of viable or necrotized tissues are suitable for a selective resection of the parenchyma, minimizing morbidity. Clinical outcome is related to a safe and effective management of inflammatory and infectious processes.
Collapse
Affiliation(s)
- Carlos Sánchez Fernández
- Department of Neurosurgery, University Clinical Hospital of Valladolid, Ramón y Cajal 3, St., 47005, Valladolid, Valladolid, Spain.
| | | | - Mónica Rivero-Garvía
- Department of Neurosurgery, University Hospital Vírgen del Rocío, Sevilla, Sevilla, Spain
| | | | - Javier Márquez Rivas
- Department of Neurosurgery, University Hospital Vírgen del Rocío, Sevilla, Sevilla, Spain
- Center for Advanced Neurology, Sevilla, Spain
| |
Collapse
|
17
|
Höhne J, Acerbi F, Falco J, Akçakaya MO, Schmidt NO, Kiris T, de Laurentis C, Ferroli P, Broggi M, Schebesch KM. Lighting Up the Tumor-Fluorescein-Guided Resection of Gangliogliomas. J Clin Med 2020; 9:jcm9082405. [PMID: 32731376 PMCID: PMC7465830 DOI: 10.3390/jcm9082405] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 01/13/2023] Open
Abstract
(1) Background: Gangliogliomas comprise a small number of brain tumors. They usually present as World Health Organization (WHO) grade I, and they delineate on gadolinium-enhanced MRI; the surgical goal is wide radical resection, and the course thereafter is usually benign. Fluorescein sodium (FL) tends to accumulate in areas with altered blood–brain barrier (BBB). Thus far, the results provided by prospective and retrospective studies show that the utilization of this fluorophore may be associated with better visualization and improvement of resection for several tumors of the central nervous system. In this study, we retrospectively studied the effect of fluorescein sodium on visualization and resection of gangliogliomas. (2) Methods: Surgical databases in three neurosurgical departments (Regensburg University Hospital; Besta Institute, Milano, Italy; and Liv Hospital, Istanbul, Turkey), with approval by the local ethics committee, were retrospectively reviewed to find gangliogliomas surgically removed by a fluorescein-guided technique by the aid of a dedicated filter on the surgical microscope from April 2014 to February 2020. Eighteen patients (13 women, 5 men; mean age 22.9 years, range 3 to 78 years) underwent surgical treatment for gangliogliomas during 19 operations. Fluorescein was intravenously injected (5 mg/kg) after general anesthesia induction. Tumors were removed using a microsurgical technique with the YELLOW 560 Filter (YE560) (KINEVO/PENTERO 900, Carl Zeiss Meditec, Oberkochen, Germany). (3) Results: No side effects related to fluorescein occurred. In all tumors, contrast enhancement on preoperative MRI correlated with bright yellow fluorescence during the surgical procedure (17 gangliogliomas WHO grade I, 1 ganglioglioma WHO grade II). Fluorescein was considered helpful by the operating surgeon in distinguishing tumors from viable tissue in all cases (100%). Biopsy was intended in two operations, and subtotal resection was intended in one operation. In all other operations considered preoperatively eligible, gross total resection (GTR) was achieved in 12 out of 16 (75%) instances. (4) Conclusions: The use of FL and YE560 is a readily available method for safe fluorescence-guided tumor resection, possibly visualizing tumor margins intraoperatively similar to contrast enhancement in T1-weighted MRI. Our data suggested a positive effect of fluorescein-guided surgery on intraoperative visualization and extent of resection during resection of gangliogliomas.
Collapse
Affiliation(s)
- Julius Höhne
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany; (N.O.S.); (K.-M.S.)
- Correspondence: ; Tel.: +49-941-944-19007
| | - Francesco Acerbi
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (F.A.); (J.F.); (C.d.L.); (P.F.); (M.B.)
| | - Jacopo Falco
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (F.A.); (J.F.); (C.d.L.); (P.F.); (M.B.)
| | - Mehmet Osman Akçakaya
- Department of Neurosurgery Liv Hospital Ulus Affiliated with Istinye University Medical Faculty, Istanbul 34340, Turkey; (M.O.A.); (T.K.)
| | - Nils Ole Schmidt
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany; (N.O.S.); (K.-M.S.)
| | - Talat Kiris
- Department of Neurosurgery Liv Hospital Ulus Affiliated with Istinye University Medical Faculty, Istanbul 34340, Turkey; (M.O.A.); (T.K.)
| | - Camilla de Laurentis
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (F.A.); (J.F.); (C.d.L.); (P.F.); (M.B.)
| | - Paolo Ferroli
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (F.A.); (J.F.); (C.d.L.); (P.F.); (M.B.)
| | - Morgan Broggi
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (F.A.); (J.F.); (C.d.L.); (P.F.); (M.B.)
| | - Karl-Michael Schebesch
- Department of Neurosurgery, University Medical Center Regensburg, 93053 Regensburg, Germany; (N.O.S.); (K.-M.S.)
| |
Collapse
|
18
|
Romano-Feinholz S, Alcocer-Barradas V, Benítez-Gasca A, Martínez-de la Maza E, Valencia-Ramos C, Gómez-Amador JL. Hybrid fluorescein-guided surgery for pituitary adenoma resection: a pilot study. J Neurosurg 2020; 132:1490-1498. [PMID: 30952130 DOI: 10.3171/2019.1.jns181512] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 01/03/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The authors conducted a pilot study on hybrid fluorescein-guided surgery for pituitary adenoma resection and herein describe the feasibility and safety of this technique. METHODS In this pilot study, the authors included all consecutive patients presenting with pituitary adenomas, functioning and nonfunctioning. They performed a hybrid fluorescein-guided surgical technique for tumor resection. An endonasal endoscopic approach was used; after exposure of the rostrum of the sphenoid sinus, they administered a bolus of 8 mg/kg of fluorescein sodium (FNa) intravenously, and during resection, they alternated between endoscopic and microscopic techniques to guide the resection under a YELLOW 560 filter. RESULTS The study included 15 patients, 7 men (47%) and 8 women (53%). Of the pituitary adenomas, 7 (46%) were nonfunctioning, 6 (40%) were GH secreting, 1 (7%) was prolactin secreting, and 1 (7%) was ACTH secreting. There were no FNa-related complications (anaphylactic reactions); yellowish staining of urine, skin, and mucosa was seen in all patients and resolved in a maximum time of 24 hours. After color spectrophotometric analysis, the authors identified a statistical difference in fluorescence among tumor, gland, and scar tissue (p = 0.01). CONCLUSIONS This is the first study of its kind to describe the feasibility and safety of using FNa to guide the resection of pituitary adenomas. The authors found this technique to be safe and feasible. It may be used to obtain better surgical results, especially for hormone-producing and recurring tumors, as well as for reducing the learning curve in pituitary adenoma surgery.
Collapse
|
19
|
Dijkstra BM, Jeltema HRJR, Kruijff S, Groen RJM. The application of fluorescence techniques in meningioma surgery-a review. Neurosurg Rev 2019; 42:799-809. [PMID: 30519770 PMCID: PMC6821664 DOI: 10.1007/s10143-018-01062-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/11/2018] [Accepted: 11/23/2018] [Indexed: 12/27/2022]
Abstract
Surgical resections of meningiomas, the most common intracranial tumor in adults, can only be curative if radical resection is achieved. Potentially, the extent of resection could be improved, especially in complex and/or high-grade meningiomas by fluorescence-guided surgery using 5-aminolevulinic acid (5-ALA), indocyanine green (ICG), or fluorescein. This review aims to summarize and evaluate these fluorescence-guided meningioma surgery techniques. PubMed and Embase were searched for relevant articles. Additionally, we checked reference lists for further studies. Forty-eight articles were included in the final analysis. 5-ALA fluoresced with varying sensitivity and selectivity in meningiomas and in invaded bone and dura mater. Although ICG was mainly applied for video angiography, one report shows tumor fluorescence 18-28 h post-ICG injection. Lastly, the use of fluorescein could aid in the identification of tumor remnants; however, detection of dural tail is highly questionable. Fluorescence-guided meningioma surgery should be a reliable, highly specific, and sensitive technique. Despite numerous studies reporting the use of fluorescent dyes, currently, there is no evidence that these tools improve the radical resection rate and long-term recurrence-free outcome in meningioma surgery without neurological deficits. Evidence regarding the effectiveness and increased safety of resection after the application of these fluorophores is currently lacking. Future research should focus on the development of a meningioma-targeted, highly sensitive, and specific fluorophore.
Collapse
Affiliation(s)
- Bianca M Dijkstra
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Hanne-Rinck J R Jeltema
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Schelto Kruijff
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rob J M Groen
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
20
|
Schebesch KM, Rosengarth K, Brawanski A, Proescholdt M, Wendl C, Höhne J, Ott C, Lamecker H, Doenitz C. Clinical Benefits of Combining Different Visualization Modalities in Neurosurgery. Front Surg 2019; 6:56. [PMID: 31632980 PMCID: PMC6781653 DOI: 10.3389/fsurg.2019.00056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 09/04/2019] [Indexed: 12/30/2022] Open
Abstract
The prevailing philosophy in oncologic neurosurgery, has shifted from maximally invasive resection to the preservation of neurologic function. The foundation of safe surgery is the multifaceted visualization of the target region and the surrounding eloquent tissue. Recent advancements in pre-operative and intraoperative visualization modalities have changed the face of modern neurosurgery. Metabolic and functional data can be integrated into intraoperative guidance software, and fluorescent dyes under dedicated filters can potentially visualize patterns of blood flow and better define tumor borders or isolated tumor foci. High definition endoscopes enable the depiction of tiny vessels and tumor extension to the ventricles or skull base. Fluorescein sodium-based confocal endomicroscopy, which is under scientific evaluation, may further enhance the neurosurgical armamentarium. We aim to present our institutional workup of combining different neuroimaging modalities for surgical neuro-oncological procedures. This institutional algorithm (IA) was the basis of the recent publication by Haj et al. describing outcome and survival data of consecutive patients with high grade glioma (HGG) before and after the introduction of our Neuro-Oncology Center.
Collapse
Affiliation(s)
| | - Katharina Rosengarth
- Department of Neurosurgery, University Medical Center Regensburg, Regensburg, Germany
| | - Alexander Brawanski
- Department of Neurosurgery, University Medical Center Regensburg, Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, University Medical Center Regensburg, Regensburg, Germany
| | - Christina Wendl
- Department of Radiology, University Medical Center Regensburg, Regensburg, Germany
| | - Julius Höhne
- Department of Neurosurgery, University Medical Center Regensburg, Regensburg, Germany
| | - Christian Ott
- Department of Neurosurgery, University Medical Center Regensburg, Regensburg, Germany
| | | | - Christian Doenitz
- Department of Neurosurgery, University Medical Center Regensburg, Regensburg, Germany
| |
Collapse
|
21
|
Osman H, Elsahy D, Slivova V, Thompson C, Georges J, Yocom S, Cohen-Gadol AA. Neurosurgical Flexible Probe Microscopy with Enhanced Architectural and Cytological Detail. World Neurosurg 2019; 128:e929-e937. [PMID: 31100529 DOI: 10.1016/j.wneu.2019.05.039] [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: 01/16/2019] [Revised: 05/03/2019] [Accepted: 05/04/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Microscopic delineation and clearance of tumor cells at neurosurgical excision margins potentially reduce tumor recurrence and increase patient survival. Probe-based in vivo fluorescence microscopy technologies are promising for neurosurgical in vivo microscopy. OBJECTIVE We sought to demonstrate a flexible fiberoptic epifluorescence microscope capable of enhanced architectural and cytological imaging for in vivo microscopy during neurosurgical procedures. METHODS Eighteen specimens were procured from neurosurgical procedures. These specimens were stained with acridine orange and imaged with a 3-dimensional (3D)-printed epifluorescent microscope that incorporates a flexible fiberoptic probe. Still images and video sequence frames were processed using frame alignment, signal projection, and pseudo-coloring, resulting in resolution enhancement and an increased field of view. RESULTS Images produced displayed good nuclear contrast and architectural detail. Grade 1 meningiomas demonstrated 3D chords and whorls. Low-grade meningothelial nuclei showed streaming and displayed regularity in size, shape, and distribution. Oligodendrogliomas showed regular round nuclei and a variably staining background. Glioblastomas showed high degrees of nuclear pleomorphism and disarray. Mitoses, vascular proliferation, and necrosis were evident. CONCLUSIONS We demonstrate the utility of a 3D-printed, flexible probe microscope for high-resolution microscopic imaging with increased architectural detail. Enhanced in vivo imaging using this device may improve our ability to detect and decrease microscopic tumor burden at excision margins during neurosurgical procedures.
Collapse
Affiliation(s)
- Hany Osman
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Deena Elsahy
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Veronika Slivova
- Enterprise Clinical Research Operations Biorepository, Indiana University Health, Indianapolis, Indiana, USA
| | - Corey Thompson
- Enterprise Clinical Research Operations Biorepository, Indiana University Health, Indianapolis, Indiana, USA
| | - Joseph Georges
- Department of Neurosurgery, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | - Steven Yocom
- Department of Neurosurgery, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | - Aaron A Cohen-Gadol
- Goodman Campbell Brain and Spine and Indiana University Department of Neurological Surgery, Indianapolis, Indiana, USA.
| |
Collapse
|
22
|
Linsler S, Ketter R, Oertel J, Urbschat S. Fluorescence imaging of meningioma cells with somatostatin receptor ligands: an in vitro study. Acta Neurochir (Wien) 2019; 161:1017-1024. [PMID: 30877475 DOI: 10.1007/s00701-019-03872-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND The use of five-aminolevulinic acid (5-ALA) in the staining of malignant glioma cells has significantly improved intraoperative radicality in the resection of gliomas in the last decade. Currently, there is no comparable selective fluorescent substance available for meningiomas. There is however a demand for intraoperative fluorescent identification of, e.g., invasive skull base meningiomas to help improve safe radical resection. Meningiomas show high expression of the somatostatin receptor type 2, offering the possibility of receptor-targeted imaging. The authors used a somatostatin receptor-labeled fluorescence dye in the identification of meningiomas in vitro. The aim of this study was to evaluate the possibility of selective identification of meningioma cells with fluorescent techniques. METHODS Twenty-four primary human meningioma cell cultures were analyzed. The tumor cells were incubated with FAM-TOC (5,6-Carboxyfluoresceine-Tyr3-Octreotide). As a negative control, four human dura tissues were cultured as well as a mixed cell culture in vitro and incubated with the same somatostatin receptor-labeled fluorescence substance. After incubation, fluorescence signal and intensity in all cell cultures were analyzed at three different time points using a fluorescence microscope with 488 nm epi-illumination. RESULTS Sixteen WHO I, six WHO II, two WHO III meningioma primary cell cultures, and four dura cell cultures were analyzed. Fluorescence was detected in all meningioma cell cultures (22 cell culture stained strongly, 2 cell cultures moderately) directly after incubation up until 4 h later. There were no differences in the quality and quantity of fluorescence signal between the various meningioma grades. The fluorescence signal persisted unchanged during the analyzed period. In the negative control, dura cell cultures remained unstained. CONCLUSIONS This study demonstrates the use of FAM-TOC in the selective fluorescent identification of meningioma cells in vitro. Further evaluation of the chemical kinetics of the applied somatostatin receptor ligand and fluorescence dye is warranted. As a next step, an experimental animal model is needed to evaluate these promising results in vivo.
Collapse
|
23
|
Cho SS, Salinas R, Lee JYK. Indocyanine-Green for Fluorescence-Guided Surgery of Brain Tumors: Evidence, Techniques, and Practical Experience. Front Surg 2019; 6:11. [PMID: 30915339 PMCID: PMC6422908 DOI: 10.3389/fsurg.2019.00011] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 02/19/2019] [Indexed: 01/10/2023] Open
Abstract
The primary treatment for brain tumors often involves surgical resection for diagnosis, relief of mass effect, and prolonged survival. In neurosurgery, it is of utmost importance to achieve maximal safe resection while minimizing iatrogenic neurologic deficit. Thus, neurosurgeons often rely on extra tools in the operating room, such as neuronavigation, intraoperative magnetic resonance imaging, and/or intraoperative rapid pathology. However, these tools can be expensive, not readily available, time-consuming, and/or inaccurate. Recently, fluorescence-guided surgery has emerged as a cost-effective method to accurately visualize neoplastic areas in real-time to guide resection. Currently, 5-aminolevulinic-acid (5-ALA) remains the only fluorophore that has been approved specifically for fluorescence-guided tumor resection. Its use has demonstrated improved resection rates and prolonged progression-free survival. However, protoporphyrin-IX, the metabolic product of 5-ALA that accumulates in neoplastic cells, fluoresces in the visible-light range, which suffers from limited tissue penetration and significant auto-fluorescence. Near-infrared fluorescence, on the other hand, overcomes these problems with ease. Since 2012, researchers at our institution have developed a novel technique using indocyanine-green, which is a well-known near-infrared fluorophore used traditionally for angiography. This Second-Window-ICG (SWIG) technique takes advantage of the increased endothelial permeability in peritumoral tissue, which allows indocyanine-green to accumulate in these areas for intraoperative visualization of the tumor. SWIG has demonstrated utility in gliomas, meningiomas, metastases, pituitary adenomas, chordomas, and craniopharyngiomas. The main benefits of SWIG stem from its highly sensitive detection of neoplastic tissue in a wide variety of intracranial pathologies in real-time, which can help neurosurgeons both during surgical resections and in stereotactic biopsies. In this review of this novel technique, we summarize the development and mechanism of action of SWIG, provide evidence for its benefits, and discuss its limitations. Finally, for those interested in near-infrared fluorescence-guided surgery, we provide suggestions for maximizing the benefits while minimizing the limitations of SWIG based on our own experience thus far.
Collapse
Affiliation(s)
- Steve S Cho
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Ryan Salinas
- Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - John Y K Lee
- Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
24
|
Göker B, Kırış T. Sodium fluorescein-guided brain tumor surgery under the YELLOW-560-nm surgical microscope filter in pediatric age group: feasibility and preliminary results. Childs Nerv Syst 2019; 35:429-435. [PMID: 30610485 DOI: 10.1007/s00381-018-04037-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 12/17/2018] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To evaluate the feasibility and safety of sodium fluorescein (Na-Fl)-guided surgery with the use of the PENTERO 900 surgical microscope (Carl Zeiss, Meditec, Oberkochen, Germany) equipped with the YELLOW-560-nm filter and low-dose Na-Fl (2 mg/kg) in pediatric brain tumor surgery. METHODS The study included 23 pediatric patients with various intracranial pathologies, who underwent Na-Fl-guided surgery between April 2015 and February 2018. Clinical features, surgical observations, extent of resection, and tumor histopathology were retrospectively analyzed. The use of YELLOW-560-nm filter was found "helpful" if the discrimination of the pinkish brain tissue and bright yellow stained tumor tissue was clear. Otherwise, it was described as "not helpful." RESULTS There were 11 female and 12 male patients with a mean age of 9.4 years. There were 7 brain stem/tectal plate gliomas, 6 supratentorial tumors, 4 intraventricular tumors, 2 pineal tumors, 2 infratentorial tumors, 1 clivus tumor, and 1 tumor with supra- and infratentorial extensions in the current series. Na-Fl was found helpful by means of the tumor demarcation in 20 instances (87%). In 11 of these 20 operations (55%), a total resection was achieved regardless of the tumor pathology. A subtotal resection was achieved in the remaining 9 patients (45%). No adverse events or side effects were encountered with regard to Na-Fl use. CONCLUSION Na-Fl guidance with the use of the YELLOW-560 filter is safe and effective during brain tumor surgery in pediatric age group.
Collapse
Affiliation(s)
- Burcu Göker
- Department of Neurosurgery, Liv Hospital Ulus, Istanbul, Turkey. .,Department of Neurosurgery, Istinye University Medical Faculty, Istanbul, Turkey.
| | - Talat Kırış
- Department of Neurosurgery, Liv Hospital Ulus, Istanbul, Turkey.,Department of Neurosurgery, Istinye University Medical Faculty, Istanbul, Turkey
| |
Collapse
|
25
|
Dijkstra BM, Motekallemi A, den Dunnen WFA, Jeltema JR, van Dam GM, Kruyt FAE, Groen RJM. SSTR-2 as a potential tumour-specific marker for fluorescence-guided meningioma surgery. Acta Neurochir (Wien) 2018; 160:1539-1546. [PMID: 29858948 PMCID: PMC6060877 DOI: 10.1007/s00701-018-3575-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/23/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Meningiomas are the most frequently occurring primary intracranial tumours in adults. Surgical removal can only be curative by complete resection; however surgical access can be challenging due to anatomical localization and local invasion of bone and soft tissues. Several intraoperative techniques have been tried to improve surgical resection, including intraoperative fluorescence guided imaging; however, no meningioma-specific (fluorescent) targeting has been developed yet. Here, we aimed to identify the most promising biomarkers for targeted intra-operative fluorescence guided meningioma surgery. METHODS One hundred forty-eight meningioma specimens representing all meningioma grades were analysed using immunohistochemistry (IHC) on tissue microarrays (TMAs) to determine expression patterns of meningioma biomarkers epithelial membrane antigen (EMA), platelet-derived growth factor β (PDGF-β), vascular endothelial growth factor α (VEGF-α), and somatostatin receptor type 2 (SSTR-2). Subsequently, the most promising biomarker was selected based on TArget Selection Criteria (TASC). Marker expression was examined by IHC in 3D cell culture models generated from freshly resected tumour material. RESULTS TMA-IHC showed strongest staining for SSTR-2. All cases were positive, with 51.4% strong/diffuse, 30.4% moderate/diffuse and only 18.2% focal/weak staining patterns. All tested biomarkers showed at least weak positivity in all meningiomas, regardless of WHO grade. TASC analysis showed that SSTR-2 was the most promising target for fluorescence guided imaging, with a total score of 21 (out of 22). SSTR-2 expression was determined on original patient tumours and 3D cultures of three established cultures. CONCLUSIONS SSTR-2 expression was highly sensitive and specific in all 148 meningiomas, regardless of WHO grade. According to TASC analysis, SSTR-2 is the most promising receptor for meningioma targeting. After establishing in vitro meningioma models, SSTR-2 cell membrane expression was confirmed in two of three meningioma cultures as well. This indicates that specific fluorescence in an experimental setting can be performed for the further development of targeted fluorescence guided meningioma surgery and near-infrared fluorescent tracers targeting SSTR-2.
Collapse
Affiliation(s)
- B M Dijkstra
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands
| | - A Motekallemi
- Department of Neurosurgery, University Medical Center Münster, Münster, Germany
| | - W F A den Dunnen
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J R Jeltema
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands
| | - G M van Dam
- Department of Surgery, Nuclear Medicine and Molecular Imaging and Intensive Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - F A E Kruyt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - R J M Groen
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
26
|
Cavallo C, De Laurentis C, Vetrano IG, Falco J, Broggi M, Schiariti M, Ferroli P, Acerbi F. The utilization of fluorescein in brain tumor surgery: a systematic review. J Neurosurg Sci 2018; 62:690-703. [PMID: 29790725 DOI: 10.23736/s0390-5616.18.04480-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Sodium fluorescein (SF) is a green, water-soluble dye with the capacity to accumulate in cerebral areas as a result of damaged blood-brain barrier (BBB); this property allows SF to concentrate specifically at the tumor site of various types of brain neoplasms, making the tumor tissue more clearly visible. EVIDENCE ACQUISITION A literature search (1947-2018) was conducted with the keywords "fluorescein neurosurgery," "YELLOW neurosurgery," "fluorescein brain tumor," "YELLOW brain tumor." We included clinical studies, clinical trials, observational studies, only conducted on humans and concerning surgery; in addition, we have included 3 articles derived from the analysis of the references of other papers. Ultimately, 57 articles were included for further analysis. EVIDENCE SYNTHESIS Fluorescein as a fluorescent tracer in neuro-oncology is gaining a wider acceptance in the neurosurgical literature: until February 1st, 2018, at least 1099 neuro-oncological patients have been operated through fluorescein-assistance, mostly only after 2012. The most important application remains the aim to improve tumor visualization and extent of resection for high-grade gliomas (HGG), but the nonspecific mechanism of action is the theoretical base for its use also for tumors different from HGG. Nevertheless, no homogenous protocol of fluorescein utilization in neurosurgical oncology can be found in literature. CONCLUSIONS Fluorescein-guided surgery is a safe and effective technique to improve visualization and resection of different CNS tumors and conditions, based on BBB alteration, with a growing evidence-based background.
Collapse
Affiliation(s)
- Claudio Cavallo
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph Hospital and Medical Center, Phoenix, AZ, USA
| | - Camilla De Laurentis
- Neurosurgical Unit II, Department of Neurosurgery, IRCCS "Istituto Neurologico C. Besta" Foundation, Milan, Italy
| | - Ignazio G Vetrano
- Neurosurgical Unit II, Department of Neurosurgery, IRCCS "Istituto Neurologico C. Besta" Foundation, Milan, Italy
| | - Jacopo Falco
- Neurosurgical Unit II, Department of Neurosurgery, IRCCS "Istituto Neurologico C. Besta" Foundation, Milan, Italy
| | - Morgan Broggi
- Neurosurgical Unit II, Department of Neurosurgery, IRCCS "Istituto Neurologico C. Besta" Foundation, Milan, Italy
| | - Marco Schiariti
- Neurosurgical Unit II, Department of Neurosurgery, IRCCS "Istituto Neurologico C. Besta" Foundation, Milan, Italy
| | - Paolo Ferroli
- Neurosurgical Unit II, Department of Neurosurgery, IRCCS "Istituto Neurologico C. Besta" Foundation, Milan, Italy
| | - Francesco Acerbi
- Neurosurgical Unit II, Department of Neurosurgery, IRCCS "Istituto Neurologico C. Besta" Foundation, Milan, Italy -
| |
Collapse
|
27
|
Belykh E, Miller EJ, Patel AA, Yazdanabadi MI, Martirosyan NL, Yağmurlu K, Bozkurt B, Byvaltsev VA, Eschbacher JM, Nakaji P, Preul MC. Diagnostic Accuracy of a Confocal Laser Endomicroscope for In Vivo Differentiation Between Normal Injured And Tumor Tissue During Fluorescein-Guided Glioma Resection: Laboratory Investigation. World Neurosurg 2018; 115:e337-e348. [PMID: 29673821 DOI: 10.1016/j.wneu.2018.04.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/06/2018] [Accepted: 04/07/2018] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Glioma resection with fluorescein sodium (FNa) guidance has a potential drawback of nonspecific leakage of FNa from nontumor areas with a compromised blood-brain barrier. We investigated the diagnostic accuracy of in vivo confocal laser endomicroscopy (CLE) after FNa administration to differentiate normal brain, injured normal brain, and tumor tissue in an animal glioma model. METHODS GL261-Luc2 gliomas in C57BL/6 mice were used as a brain tumor model. CLE images of normal, injured normal, and tumor brain tissues were collected after intravenous FNa administration. Correlative sections stained with hematoxylin and eosin were taken at the same sites. A set of 40 CLE images was given to 1 neuropathologist and 3 neurosurgeons to assess diagnostic accuracy and rate image quality (1-10 scale). Additionally, we developed a deep convolution neural network (DCNN) model for automatic image classification. RESULTS The mean observer accuracy for correct diagnosis of glioma compared with either injured or uninjured brain using CLE images was 85%, and the DCNN model accuracy was 80%. For differentiation of tumor from nontumor tissue, the experts' mean accuracy, specificity, and sensitivity were 90%, 86%, and 96%, respectively, with high interobserver agreement overall (Cohen κ = 0.74). The percentage of correctly identified images was significantly higher for images with a quality rating >5 (104/116, 90%) than for images with a quality rating ≤5 (32/44, 73%) (P = 0.007). CONCLUSIONS With sufficient FNa present in tissues, CLE was an effective tool for intraoperative differentiation among normal, injured normal, and tumor brain tissue. Clinical studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Evgenii Belykh
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA; Irkutsk State Medical University, Irkutsk, Russia
| | - Eric J Miller
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Arpan A Patel
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | | | - Nikolay L Martirosyan
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Kaan Yağmurlu
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Baran Bozkurt
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | | | - Jennifer M Eschbacher
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Peter Nakaji
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Mark C Preul
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA.
| |
Collapse
|
28
|
Akçakaya MO, Göker B, Kasımcan MÖ, Hamamcıoğlu MK, Kırış T. In Reply to the Letter to the Editor "Fluorescein Sodium in Intracranial Meningioma Surgery". World Neurosurg 2017; 108:968. [PMID: 29179416 DOI: 10.1016/j.wneu.2017.09.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 09/09/2017] [Indexed: 11/25/2022]
Affiliation(s)
| | - Burcu Göker
- Department of Neurosurgery, Liv Hospital, Istanbul/Turkey
| | | | | | - Talat Kırış
- Department of Neurosurgery, Liv Hospital, Istanbul/Turkey
| |
Collapse
|
29
|
Schebesch KM, Brawanski A, Höhne J. Fluorescein Sodium in Intracranial Meningioma Surgery. World Neurosurg 2017; 108:967. [PMID: 29179415 DOI: 10.1016/j.wneu.2017.08.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 08/08/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Karl-Michael Schebesch
- Department of Neurosurgery and Wilhelm Sander-Neuro-Oncology Unit, University Med, Regensburg, Germany.
| | - Alexander Brawanski
- Department of Neurosurgery and Wilhelm Sander-Neuro-Oncology Unit, University Med, Regensburg, Germany
| | - Julius Höhne
- Department of Neurosurgery and Wilhelm Sander-Neuro-Oncology Unit, University Med, Regensburg, Germany
| |
Collapse
|